Handbook Of: Experimental Pharmacology
Handbook Of: Experimental Pharmacology
Experimental Pharmacology
Volume 122
Editorial Board
G.V.R.Born,London
P. Cuatrecasas, Ann Arbor, MI
D. Ganten, Berlin
H. Herken, Berlin
K.L. Melmon, Stanford, CA
Springer
Berlin
Heidelberg
New York
Barcelona
Budapest
Hong Kong
London
Milan
Paris
Santa Clara
Singapore
Tokyo
Mechanisms
of Drug Interactions
Contributors
G.L. Amidon, H.H. Blume, J.R. erison, P.F. D'Arcy
P.A.G.M. De Smet, J.D. Ferrero, J.P. Griffin, e.M. Hughes
E. Lipka, J.e. McElnay, M. Schorderet, B.S. Schug, A. Somogyi
P.G. Welling, S. Yosselson-Superstine
Editors
P.F. D'Arcy, J.C. McElnay and P.G. Welling
Springer
ISBN-13: 978-3-642-64658-4 e-ISBN-13: 978-3-642-61015-8
DOI: 10.107/978-3-642-61015-8
Over the years a number of excellent books have classified and detailed drug-
drug interactions into their respective categories, e.g. interactions at plasma
protein binding sites; those altering intestinal absorption or bioavailability;
those involving hepatic metabolising enzymes; those involving competition or
antagonism for receptor sites, and drug interactions modifying excretory
mechanisms. Such books have presented extensive tables of interactions and
their management. Although of considerable value to clinicians, such publica-
tions have not, however, been so expressive about the individual mechanisms
that underlie these interactions.
It is within this sphere of "mechanisms" that this present volume
specialises. It deals with mechanisms of in vitro and in vivo, drug-drug, drug-
food and drug-herbals interactions and those that cause drugs to interfere with
diagnostic laboratory tests. We believe that an explanation of the mechanisms
of such interactions will enable practitioners to understand more fully the
nature of the interactions and thus enable them to manage better their clinical
outcome.
If mechanisms of interactions are better understood, then it may be pos-
sible for the researcher to develop meaningful animal/biochemical/tissue cul-
ture or physicochemical models to which new molecules could be exposed
during their development stages. The present position, which largely relies on
patients experiencing adverse interactions before they can be established or
documented, can hardly be regarded as satisfactory.
This present volume is classified into two major parts; firstly, pharmacoki-
netic drug interactions and, secondly, pharmacodynamic drug interactions.
Within these parts, we have been fortunate to enlist the help of acknowledged
experts in preparing specific chapters focusing on aspects of the interaction
spectrum.
We believe that this volume will add much to that which is currently
known about drug interactions and will directly enhance the safer use of
medicines.
AMIDON, G.L., The University of Michigan, 2012 Pharmacy Bldg., Ann Arbor,
MI 48109-1065, USA
LIPKA, E., The University of Michigan, 2032 Pharmacy Bldg., Ann Arbor, MI
48109-1065, USA, and TSRL Inc., Ann Arbor, MI 48108, USA
CHAPTER 1
Introduction
P.F. D' ARCY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . .. .. 1
A. A Widening Problem. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
I. Scope of This Present Volume. . . . . . . . . . . . . . . . . . . . . . . . . . . 2
B. Pharmacokinetic Drug Interactions. . . . . . . . . . . . . . . . . . . . . . . . . . . 2
I. Drug-Drug and Nutrient-Drug Interactions
at the Absorption Site. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
II. Drug Interactions at Plasma- and Tissue-Binding Sites. . . . . . . 4
III. Drug Interactions and Drug-Metabolising Enzymes. . . . . . . . . 4
IV. Interactions Involving Renal Excretory Mechanisms. . . . . . . . 4
C. Pharmacodynamic Drug Interactions. . . . . . . . . . . . . . . . . . . . . . . . . . 5
I. Drug-Drug Interactions at the Receptor
and Other Active Sites. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
II. Synergistic Interactions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
III. Drug Interactions In Vitro. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
IV. Age and Genetic Factors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
V. Interference with Laboratory Testing. . . . . . . . . . . . . . . . . . . . . 7
VI. Herbal and Other Non-orthodox Medicines. . . . . . . . . . . . . . . . 7
D. Comment.................................................. 8
I. Drug Interactions:
Hazardous and Expensive Use of Resources? . . . . . . . . . . . . . . . 8
II. Sources of Information. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
III. The Literature on Drug Interactions. . . . . . . . . . . . . . . . . . . . . . 9
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
x Contents
CHAPTER 2
Drng Interactions in the Gastrointestinal Tract
and Their Impact on Drug Absorption and Systemic Availability:
A Mechanistic Review
E. LIPKA, J.R. CRISON, B.S. SCHUG, H.H. BLUME and G.L. AMIDON
With 9 Figures. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
A. Introduction............................................... 13
B. Physicochemical Interactions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
I. Complexation with Metal Ions. . . . . . . . . . . . . . . . . . . . . . . . . . . 18
II. Binding to Resins. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
III. Complexation with Bile Salts. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
IV. Nonspecific Adsorption . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
C. Interactions with Drugs That Influence GI Motility and pH . . . . . . 21
I. Alteration of Gastrointestinal Motility. . . . . . . . . . . . . . . . . . . . . 21
II. pH Alteration in the Gastrointestinal Tract. . . . . . . . . . . . . . . . . 26
D. Interactions Between Drugs
That Share the Same Absorption Mechanism. . . . . . . . . . . . . . . . . . . 27
I. Passive Absorption. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
II. Carrier-Mediated Absorption. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
E. Interactions as a Function of Intestinal Metabolism. . . . . . . . . . . . . 33
F. Altered Absorption as a Result
of Drug-Induced Mucosal Changes. . . . . . . . . . . . . . . . . . . . . . . . . . . 37
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
CHAPTER 3
Drug-Food Interactions Affecting Drug Absorption
P.G. WELLING. With 23 Figures. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
A. Introduction............................................... 45
I. Food. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
II. Dosage Form. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
B. Influence of Food on the Gastrointestinal Tract. . . . . . . . . . . . . . . . 46
C. Direct Effect of Food on Drug Absorption. . . . . . . . . . . . . . . . . . . . 49
D. Interactions Causing Reduced Drug Absorption. . . . . . . . . . . . . . . . 51
E. Interactions Causing Delayed Drug Absorption. . . . . . . . . . . . . . . . . 63
F. Interactions Causing Increased Drug Absorption. . . . . . . . . . . . . . . 77
G. Interactions Causing Accelerated Drug Absorption. . . . . . . . . . . . . 95
H. Cases in Which Food Has No Effect on Drug Absorption....... 98
I. Conclusions................................................ 110
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
Contents XI
CHAPTER 4
Drug Interactions at Plasma and Tissue Binding Sites
Joe. McELNAYo With 7 Figures 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 125
A. Introduction 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 125
Bo Proteins Involved in Drug Binding 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 125
e. Influence of Plasma and Tissue Binding on Drug Kinetics 0 0 0 0 0 0 0 0 127
Do Displacement of Drugs from Binding Sites 000000000000000000000 129
Eo Therapeutic Consequences
of Plasma Binding Displacement Drug-Drug Interactions 00000000 130
I. Rapid Intravenous Infusion of Displacing Agent 0 0 0 0 0 0 0 0 0 0 0 133
II. Parenteral Administration of Displaced Drug
Having High Extraction Ratio 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 134
III. Therapeutic Drug Monitoring and Drug Displacement
from Plasma Binding Sites 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 134
F. Therapeutic Consequences
of Tissue Binding Displacement Interactions 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 136
Go Displacement of Drugs from Binding Sites
by Endogenous Materials 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 138
Ho Disease States and Altered Plasma Protein Binding 0 0 0 0 0 0 0 0 0 0 0 0 0 139
I. Conclusions 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 000 0 0 0 0 0 0 0 0 0 0 0 0 0 0 145
References 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 145
CHAPTER 5
Drug Interactions and Drug-Metabolising Enzymes
PoFo D' ARCY 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 151
A. General Introduction 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 151
Bo Extrahepatic Microsomal Forms of Cytochrome P450 0 0 0 0 0 0 0 0 0 0 0 154
C. Genetic Polymorphism 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 156
Do Age and Disease and Cytochrome P450 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 000 157
Eo Clinical Importance of Enzyme Induction or Inhibition 0 0 0 0 0 0 0 0 0 0 158
I. Enzyme Inducers 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 158
II. Enzyme Inhibitors 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 160
Fo Conclusions 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 000000000000000 164
References 00000000000000000000000000000000000000000000000000 0 166
CHAPTER 6
Drug Interactions Involving Renal Excretory Mechanisms
A. SOMOGYI. With 5 Figures 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 173
Ao Introduction 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 173
B. Mechanisms of Renal Excretory Clearance 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 173
XII Contents
CHAPTER 7
Drug-Drug Interactions at Receptors and Other Active Sites
M. SCHORDERET and J.D. FERRERO. With 5 Figures. . . . . . . . . . . . . . . . . . 215
A. Introduction............................................... 215
B. Mechanisms of Pharmacodynamic Interactions. . . . . . . . . . . . . . . . . 215
I. Transmitter Systems.................................... 215
1. Noradrenergic Synapse. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
2. Dopaminergic Synapse. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 219
3. Serotonergic Synapse. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 220
4. Cholinergic Synapse. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
5. GABAergic Synapse. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
II. Ion Channels. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 224
Contents XIII
CHAPTER 8
Synergistic Drug Interactions
J.P. GRIFFIN and P.F. D'ARCY.. . ...... . ..... . . . . ....... . ...... . . 235
A. Introduction: "Mithridatium"................................ 235
B. Early Use of Probenecid with Penicillin. . . . . . . . . . . . . . . . . . . . . . . 236
e. Diuretic Combinations. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
D. Co-trimoxazole............................................ 237
E. Combination Treatment in Control of Epilepsy. . . . . . . . . . . . . . . . . 238
F. Antitubercular Drugs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 239
G. Anti-Leprosy Treatments. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 240
H. Peptic Ulcer Therapy. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 240
I. Non-Insulin-Dependent (Maturity Onset) Diabetes. . . . . . . . . . . . . 242
J. Cancer Chemotherapy. .. ...... . . ..... . ....... . . ..... . .. .... 245
K. Beneficial Interactions: A Philosophical Approach. . . . . . . . . . . . . . 246
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 246
CHAPTER 9
In Vitro Drug Interactions
J.e. McELNAY and e.M. HUGHES. With 7 Figures. . . . . . . . . . . . . . . . . . 249
A. Introduction............................................... 249
B. Incompatibility Interactions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
e. In Vitro Drug Interactions with Pharmaceutical Packaging
and Intravenous Administration Equipment. . . . . . . . . . . . . . . . . . . . 252
I. General Properties of Plastics ............................ 252
II. General Properties of Glass. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 254
III. Mechanisms of Interaction with Pharmaceutical Packaging. . . 254
1. Sorption............................................ 257
XIV Contents
2. Leaching............................................ 262
3. Permeation.......................................... 264
4. Polymer Modification. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 264
IV. Drug Interactions with Contact Lenses. . . . . . . . . . . . . . . . . . . . . 265
D. In Vitro Drug Interactions in Therapeutic Drug Monitoring
and Drug Analysis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
I. Cyclosporin............................................ 265
II. Chloroquine........................................... 266
E. In Vitro Drug Interactions as a Result of Formulation Changes. . . 266
I. Problems with Sustained-Release Formulations. . . . . . . . . . . . . 268
II. Drug Excipient Interactions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 270
III. Formulation Effects on Rectal Bioavailability . . . . . . . . . . . . . . 272
F. Conclusions................................................ 274
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 274
CHAPTER 10
Age and Genetic Factors in Drug Interactions
J.e. McELNAY and P.F. D'ARCY. With 2 Figures. . ........ . . . . ...... 279
A. Introduction............................................... 279
B. Age....................................................... 279
I. Pharmacokinetics in the Elderly. . . . . . . . . . . . . . . . . . . . . . . . . . 284
1. Absorption.......................................... 284
2. Distribution......................................... 284
3. Metabolism.......................................... 287
4. Excretion........................................... 288
II. Pharmacodynamics in the Elderly. . . . . . . . . . . . . . . . . . . . . . . . . 289
III. Inappropriate, Unnecessary and Interacting Medication. . . . . . 291
IV. Logistics: Age and Medicine Consumption. . . . . . . . . . . . . . . . . 293
V. Long-Term Treatments. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293
e. Genetic Factors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 294
D. Comment................................................. 300
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 301
CHAPTER 11
Drugs Causing Interference with Laboratory Tests
S. YOSSELSON-SUPERSTINE. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
A. Introduction............................................... 305
B. Mechanisms of Drug-Test Interactions. . . . . . . . . . . . . . . . . . . . . . . . 306
I. PharmacologicalInterferences............................ 306
1. Effect on Glucose Determination. . . . . . . . . . . . . . . . . . . . . . 306
2. Effect on Uric Acid Determination. . . . . . . . . . . . . . . . . . . . . 306
Contents XV
3. Intramuscular Injections
and Muscle Enzyme Determination. . . . . . . . . . . . . . . . . . . . . 307
4. Effect of Drug-Drug Interactions. . . . . . . . . . . . . . . . . . . . . . . 307
II. Methodological Interferences. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 308
1. Colorimetric Interferences. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 308
2. Effect on pH of the Assay Environment. . . . . . . . . . . . . . . . . 314
3. Interference with Chromatographic Methods. . . . . . . . . . . . . 315
4. Interference with Enzymatic Reactions. . . . . . . . . . . . . . . . . . 317
5. Interference with Immunoassays. . . . . . . . . . . . . . . . . . . . . . . 319
C. Design of a Study for Evaluation of Analytical Interference. . . . . . 320
D. Conclusions................................................ 321
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 322
CHAPTER 12
Drug Interactions with Herbal and Other Non-orthodox Remedies
P.A.G.M. DE SMET and P.F. D'ARCY............................. 327
A. Introduction............................................... 327
B. Animal Agents. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 329
I. Fish Oil.... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 329
II. Chinese Toad Venom. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 329
C. Amino Acids. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 329
I. L-Tryptophan. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 329
D. Vitamins.................................................. 330
E. Minerals.................................................. 331
F. Dietary Fads. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 331
G. Herbal Drugs....... . . . . . ..... . . . .... . . . ........ . ...... . . .. 332
I. Effects of Herbal Drugs
on Orthodox Drug Pharmacokinetics:
Effects on Absorption. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 333
1. Dietary Fibres. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 333
2. Guar Gum......... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 334
3. Tannins............................................. 334
II. Effects of Herbal Drugs
on Orthodox Drug Pharmacokinetics:
Effects on Elimination. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 336
1. Cola Nut. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 336
2. Eucalyptus Species. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 336
3. Grapefruit Juice. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 336
4. Herbal Smoking Preparations. . . . . . . . . . . . . . . . . . . . . . . . . . 336
5. Kampo Medicines. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 336
6. Liquorice............................................ 337
III. Effects of Orthodox Drugs
on Herbal Drug Pharmacokinetics. . . . . . . . . . . . . . . . . . . . . . . . 338
XVI Contents
A. A Widening Problem
Over 20 years ago, an editorial on drug interactions in the Lancet said that the
"publication of huge lists and tables will induce in doctors a drug-interaction-
anxiety syndrome and lead to therapeutic paralysis". This prediction has not
come about although the problem of drug interactions is still with us and the
spectrum is widening as new drugs are introduced. Indeed it could be said that
the nature of the problem has also widened for in the intervening years drug
interactions have come to embrace interactions with food and with herbal
medicines as well as the more numerous and better recognised drug-drug
interactions.
One of the advantages of the attention that has been focused on adverse
drug interactions over the past 20 years or so is that many drug-drug and drug-
food interactions are now predictable and many of the unwanted conse-
quences of using drug combinations can be avoided by simply adjusting the
dosage of one or more of the interactants. As a result of this, there has been a
considerable improvement in the safety and efficacy of therapy with drug
combinations.
Nowadays the focus on drug interactions tends to be more on their haz-
ards than their advantages. This is probably the correct orientation since the
vast majority of interactions are hazardous. An awareness of the possible
hazards of medication and possible interactions between drugs on the part of
those who use them, doctors and other health professionals, can only result in
better therapy with benefit to the patient in terms of both safety and efficacy.
Many excellent publications have explored the nature of drug interactions and
their message has filtered down to the patient level, being expressed by the
maxim "do not use two drugs when one will do".
Whereas over the years books have classified and detailed drug-drug
interactions into their respective categories, e.g., interactions at plasma pro-
tein-binding sites; interactions altering intestinal absorption or bioavailability;
interactions involving the hepatic-metabolising enzymes; interactions involv-
ing competition or antagonism for receptor sites, and finally drug interactions
modifying excretory mechanisms, they have not been so expressive about the
individual mechanisms that underlie these interactions. Their prime objective
was to inform about the interaction and its clinical management.
2 P.F. D'ARCY
their potential frequency is far greater since food is by far the most common
substance associated with the ingestion of oral doses of medicine. In my
experience, medical and pharmacy practitioners are not well informed about
food-drug interactions and therefore may fail to pass on cautionary advice to
their patients.
Perhaps one of the most troublesome and well reported of food-drug
interactions was the interaction between tyramine-containing foods and
monoamine oxidase inhibitor (MAOI) antidepressants during the early 1960s
(see reviews by D' ARCY 1979; BROWN et al. 1989; LIPPMAN and NASH 1990).
Deaths from eating cheese whilst taking MAOIs was given great publicity and
this type of interaction did much to create an awareness of the potential of
foods to interact with medicines.
Monoamine oxidase type A preferentially oxidatively de-aminates
adrenaline, noradrenaline and serotonin, while type B preferentially
metabolises benzylamine and phenylethylamine. Dopamine and tyramine are
de-aminated by both forms of the enzyme. The traditional MAOIs, such as
phenelzine and tranylcypromine, inhibit both types A and B of the enzyme.
Selective MAOIs, such as brofaromine, moclobemide and clorgyline, have
been introduced with a selective and reversible inhibition of monoamine
oxidase (type A), and selegiline, which reversibly inhibits type B, is an adjunct
in the treatment of parkinsonism. The dietary restrictions that need to be
observed with the traditional MAOIs (type A and B) are much less stringent
with the new selective inhibitors.
It was largely the interactions with food that led to a virtual replacement
of the use of MAOI antidepressents by the tri- and polycyclic antidepressants.
That this may have been a collective error of judgement is now being realised;
perhaps the MAOIs are not so bad after all!
The effect of heavy metal ions in milk and other dairy products on the
absorption of tetracyclines was another milestone which raised the sensibility
of clinicians to the dangers of food-drug interactions (BRAYBROOKS et al. 1975;
CHIN and LACH 1975; NEUVONEN 1976).
I well recall in the late 1940s, after tetracycline had been first introduced,
advising mothers to open the capsules of tetracycline and dissolve the contents
in a little milk before giving the antibiotic to their infants. But it took almost
20 years before the milk-tetracycline interaction was recognised. Once this
interaction was realised, it was only a relatively short time before the spectrum
of such interactions was widened and it was realised that it was not uncommon
that the absorption from the gut of other antimicrobial agents, or reduc-
tion of their bioavailability, could be seriously influenced by food or metal
ions. Even more recent work has shown that the bisphosphonates, used to
treat a broad range of bone disorders, including osteolytic bone metastases,
hyperparathyroidism, Paget's disease of bone and established vertebral
osteoporosis in women, can be completely antagonised by food and vitamin-
mineral supplements with a high calcium content (FOGELMAN et al. 1984, 1985,
1986; FELS et al. 1989; COMPSTON 1994).
4 P.F. D'ARCY
D. Comment
References
Basu TV (1988) Drug nutrient interactions. Croom Helm, London
Boman G, Hanngren A, Malmborg A, Borga 0, Sjoqvist F (1971) Drug interaction:
decreased serum concentrations of rifampicin when given with PAS. Lancet
1:800
Braybrooks MP, Barry BW, Abbs ET (1975) The effect of mucin on the bioavailability
of tetracycline from the gastrointestinal tract: in vivo, in vitro correlation. J Pharm
Pharmacol 27:508---515
Brown C, Taniguchi G, Yip K (1989) The monoamine oxidase inhibitor-tyramine
interaction. J Clin Pharmacol 29:529-532
Chin TF, Lach JL (1975) Drug diffusion and availability: tetracycline metallic chela-
tion. Am J Hosp Pharm 32:625-629
Compston JE (1994) The therapeutic use of bisphosphonates. Br Med J 309:711-715
Conney AH, Miller EC, Miller JA (1956) The metabolism of methylated aminoazo
dyes. V. Evidence for induction of enzyme synthesis in the rat by 3-methylcholan-
threne. Cancer Res 16:450-459
Conney AH, Miller EC, Miller JA (1957) Substrate-induced synthesis and other
properties of benzpyrenehydroxylase in rat liver. J BioI Chern 228:753-766
D'Arcy PF (1979) Drug interactions. In: D'Arcy PF, Griffin JP (eds) Iatrogenic dis-
eases, 2nd edn. Oxford University Press, Oxford, pp 45-76
Dossetor J (1975) Drug interactions with oral contraceptives. Br Med J 4:467-468
10 P.F. D'ARCY: Introduction
Fels JP, Necciari J, Toussain P, Debry G, Luckx A, Scheen A (1989) Effect of food
intake on kinetics and bioavailability of (4-chlorophenyl) thiomethylene
bisphosphonic acid (Abstr). Calcif Tissue Int 44 [Suppl]:S-104
Fogelman I, Smith ML, Mazess R, Bevan JA (1984) Absorption of diphosphonate in
normal subjects (Abstr). Calcif Tissue Int 36 [SuppI2]:574
Fogelman I, Smith ML, Mazess R, Bevan JA (1985) Absorption of oral diphosphonate
in normal subjects (Abstr). Bone 6:54
Fogelman I, Smith ML, Mazess R, Wilson MA, Bevan JA (1986) Absorption of oral
diphosphonate in normal subjects. Clin Endocrinol (Oxf) 24:57-62
Levy G (1970) Biopharmaceutical considerations in dosage form and design. In:
Sprowls JB (ed) Prescription pharmacy, 2nd edn. Lippincott, Philadelphia, pp 70,
75, 80
Lippman SB, Nash K (1990) Monoamine oxidase inhibitor update: potential adverse
food and drug interactions. Drug Saf 5:195-204
Neuvonen PJ (1976) Interactions with the absorption of tetracyclines. Drugs 11:45-54
Neuvonen PJ, Gothoni G, Hackman R, Bj6rksten K (1970) Interference of iron with
the absorption of tetracyclines in man. Br Med J 4:532-534
Roe AR (1989) Diet and drug interactions. Van Nostrand Reinhold, New York
Trovato A, Nuhlicek DN, Midtling JE (1991) Drug-nutrient interactions. Am Fam
Physician 44:1651-1658
Tyrer JH, Eadie MJ, Sutherland JM, Hooper WD (1970) Outbreak of anticonvulsant
intoxication in an Australian city. Br Med J 2:271-273
Welling PG (1977) Influence of food and diet on gastrointestinal drug absorption. A
review. J Pharmacokin et Biopharm 5:291-334
Wharton R, Lewith G (1986) Complementary medicine and the general practitioner.
Br Med J 292:1498-1500
Section I
Pharmacokinetic Drug Interactions
CHAPTER 2
Drug Interactions in the Gastrointestinal Tract
and Their Impact on Drug Absorption and
Systemic Availability: A Mechanistic Review
E. LIpKA, l.R. CRISON, B.S. SCHUG, H.H. BLUME, and G.L. AMIDON
A. Introduction
The effect of drug interactions with other drugs, excipients and gastrointesti-
nal contents on their oral absorption and systemic availability have been
extensively investigated over the past 50 years. These interactions can be
classified as direct interactions between the drug and other drugs or compo-
nents in the formulation, e.g., excipients in the gastrointestinal tract altering
the drugs' thermodynamic activity for absorption or, alternatively, the effects
may be indirect through alteration of gastrointestinal transit, gastrointestinal
secretions, or gastrointestinallhepatic metabolism or elimination. Recently
attention has been directed to metabolism interactions in the liver and even
more recently in the gastrointestinal mucosal tissue. These interactions can
be caused by direct inhibition of enzyme metabolism or enzyme induction,
resulting in changes in the absorption rate and leading to altered oral
bioavailability. These metabolic effects also can be indirect, through the
position dependence of metabolizing enzyme levels in the gastrointestinal
tract; consequently transit changes can alter metabolic profiles and systemic
availability. Recent interest has been directed to carrier-mediated absorption,
where drug interactions can occur through competition for carrieres) respon-
sible for absorption and, finally, it has been proposed that the P-glycoprotein
exporter may be responsible for some of the observed drug interactions
and nonlinear absorption effects. This review will focus on examples of the
various types of mechanistic interactions noted above. The interaction of
drugs with foods, a very important component in drug regulation today,
is reviewed elsewhere in this volume. A more comprehensive survey of
relevant drug-drug interactions than can be covered in this review can be
found in Table 1.
B. Physicochemical Interactions
The two physicochemical interactions that affect oral absorption arise due to
complexation of the drug with opposite-charged ion species or to nonspecific
adsorption of the drug. In this section, examples of complexation of drugs with
Table 1. Summary of drug-drug interactions influencing absorption. (Modified from WELLING 1984)
p-Aminosalicylate (PAS) Diphenhydramine Delayed Delayed stomach LAVIGNE and MARCHAND (1973)
emptying
Aspirin Charcoal Reduced Adsorption NEUVONEN et al. (1978); LEVY and
TSUCHIYA (1972)
Aspirin (enteric-coated) Antacids Increased rate Faster drug release FELDMAN and CARLSTEDT (1974)
Carbamazepine Charcoal Reduced Adsorption NEUVONEN and ELONEN (1980)
Cefdinir Iron ion Reduced rate and Chelation VENO et al. (1993)
extent
Cephalexin Cholestyramine Reduced Adsorption or PARSONS and PADDOCK (1975)
steatorrhea
Chlorothiazide Colestipol Reduced Binding KAUFFMAN and AZARNOFF (1973)
Metoclopramide Decreased Absorption window OSMAN and WELLING (1983)
or dissolution
Propantheline Increased Absorption window OSMAN and WELLING (1983)
or dissolution
Chlorpromazine Antacids Reduced Adsorption FORREST et al. (1970); FANN et al.
Benzhexol Reduced Delayed stomach (1973) RIVERA-CALIMLIM et al.
(trihexphenidyl) emptying (1973)
Chlortetracycline Antacids Reduced Adsorption SEED and WILSON (1950);
GREENSPAN et al. (1951);
WAISBREN and HUECKEL (1950)
Ciprofloxacin Antacids Reduced Chelation LODE (1988) and NIX et al. (1989)
Clindamycin Kaolin-pectin Delayed Adsorption ALBERT et al. (1978b)
Diazepam Antacids Delayed Adsorption GREENBLATT et al. (1978)
Dicoumarol Magnesium hydroxide Increased Chelation AMBRE and FISCHER (1973)
(bishydroxycoumarin)
Digitoxin Cholestyramine Increased elimination Interrupted enterophepatic CALDWELL et al. (1971)
rate circulation
Digoxin Neomycin Reduced Sprue-like syndrome, LINDENBAUM et a1. (1972)
malabsorption
Antacids Reduced Adsorption and BROWN and JUHL (1976);
faster stomach ALBERT et a1. (1978a)
emptying (partly)
Charcoal Reduced Adsorption NEUYONEN et a1. (1978)
Metoclopramide Reduced Limited dissolution MANNINEN et a1. (1973)
Propantheline Increased Dissolution MANNINEN et a1. (1973)
Doxycycline Ferrous sulfate Reduced Chelation, influencing NEUYONEN and PENTTILA (1974);
absorption and NEUVONEN et a1. (1970)
elimination
Enoxacin Ranitidine Reduced Increased pH LEBSACK et a1. (1992)
Ethanol Metoclopramide Increased rate Faster stomach emptying GIBBONS and LANT (1975)
Propantheline Reduced rate Delayed stomach emptying GIBBONS and LANT (1975)
Atropine Reduced rate Delayed stomach emptying GIBBONS and LANT (1975)
Hydrochlorothiazide Propantheline Delayed, but Delayed stomach emptying BEERMAN and GROSCHINSKY-
increased GRIND (1978)
Ferrous ion Tetracycline Reduced Chelation NEUVONEN et a1. (1975); HEINRICH
et a1. (1974)
Isoniazid Antacids Delayed and reduced Adsorption and first-pass HURWITZ and SCHLOZMAN (1974)
metabolism
Levodopa Homatropine Reduced patient Delayed stomach emptying, FERMAGLICH and O'DOHERTY
response increased metabolism in (1972)
stomach
Antacids Increased Faster stomach emptying REVERA-CALIMLIM et a1. (1971)
Metoclopramide Increased rate Faster stomach emptying MORRIS et a1. (1976)
Lignocaine (lidocaine) General anesthetics Delayed Delayed stomach emptying ADJEPON-YAMOAH et a1. (1973)
Lincomycin Sodium cyclamate Reduced WAGNER (1969)
Lithium Metoclopramide Increased rate Faster stomach emptying CRAMMER et a1. (1974)
Propantheline Delayed Delayed stomach emptying CRAMMER et a1. (1974)
Methacycline Ferrous sulfate Reduced Chelation NEUVONEN et a1. (1970)
Nadolol Trihydroxy bile saits, Reduced Poorly soluble micelle YAMAGUCHI (1986)
sodium cholate formation
Table 1. Continued
metal ions, resins and bile salts as well as adsorption of drugs on charcoal will
be discussed.
(ug/ml)
e max 1.71 ± 0.42 0.16 ± 0.14 1.28 ± 0.23
(h)
{max 4.2 ± 1.0 1.8 ± 2.1 3.3 ± 0.5
AUCO-12 (ugh/ml) 10.3 ± 1.35*'** 0.78 ± 0.25 6.55 ± 1.61
AUC0-3 (jigh/ml) 2.13 ± 0.83*** 0.38 ± 0.3 1.95 ± 0.24
AUC3-12 (ugh/ml) 8.03 ± 1.72*,** 0.4 ± 0.31 4.60 ± 1.54
Data are mean values ± SD, emaX" maximum plasma concentration; {maX' time to reach
e max ; AUC, area under the plasma concentration-time curve. Study 1, cefdinir alone;
study 2, cefdinir and iron ion preparation (simultaneous administration); study 3, iron
preparation 3 h after cefdinir administration.
* p < 0,01, study 1 versus study 2; ** P < 0.05, study 1 versus study 3; *** P < 0,05, study
1 versus study 2.
Fig. 1. Structures of cefdinir and the formation of cefdinir-iron ion complex. The table
summarizes the pharmacokinetic parameters for cefdinir. (Adapted from UENO et al.
1993)
Drug Interactions in the Gastrointestinal Tract 19
•
~
20
!
...t
~
III
QI
~
c:
...t
....
....III 10 4)
e
III
4)
4)
~ 4)
4)
QI
-=Ilo
~
0
tl
0 10 20 40
Concentration of bile salts (nut)
Fig. 2. Effect of bile salts on nadolol absorption 4 h after injection into the rat jejunum
loop. (Adapted from YAMAGUCHI et al. 1986b). closed circles, sodium cholate; closed
squares, sodium glycocholate; closed triangles sodium taurocholate; open circles, so-
dium chenodeoxycholate; open squares, sodium deoxycholate; open triangles, sodium
lithocholate. Dose of nadolol was 0.01 mmol/rat, and that of bile salts was 0.005-
O.lmmollrat. Each point represents the mean value for at least four rats. a) P < 0.01
compared to the control (without bile salts)
Drug Interactions in the Gastrointestinal Tract 21
D n -- d·1SS0lutlOn DC
· num b er -- - _s. -4Jrr~ I
- - . tres -- tres . 3DC s pro2
ro 4 3
-Jrr
3 0P
t Diss = -r~-P
- =.tIme reqUIre
. d f or a partIe
. 1e to d·1SS01ve
3DC s
1
t~bs = k abs = ( S /V) Peff = 2 xRPeff = the effectIve
. absorptlOn
. rate constant
It is clear from the definition of the dissolution number that if the dissolution
rate and absorption rate in the intestine are constant (constant permeability,
24 E. LIPKA et al.
1.0
:f 0. 5
0.25
E 0.15
c ~
.;(0 8....
elli
=a- .~ 0.10
LI-'a
F '" 0.06
0.05
0.00 +---....---....---....---....----.
o 10 20 30 40 50
Percent Deviation from Mean SITT
Fig. 4. Predicted fraction dose absorbed for lOO-.um particles of digoxin at different
small intestinal transit times (SlIT)
Treatment Cmax (Jig/ml) t max (h) AVC (ugh/ml) t1l2 (h) Cl, (ml/min)
Enoxacin alone 2.74 ± 0.68 1.3 ± 0.5 16.1 ± 3.6 5.2 ± 1.0 182 ± 73
Enoxacin + 1.70 ± 0.66* 2.0 ± 1.0 11.9 ± 5.2* 5.7 ± 1.0* 211 ± 46
ranitidine
Enoxacin + 2.49 ± 0.55 2.1 ± 0.9 16.2 ± 4.9 5.2 ± 0.8 198 ± 81
pentagastrin
Enoxacin + 2.93 ± 0.66 1.5 ± 0.5 17.7±5.6 5.5 ± 0.9* 212 ± 86
ranitidine +
pentagastrin
Cmax> peak plasma concentration; tmaX' time to reach peak concentration; AVC, area
under the plasma concentration-time curve; (liZ, half-life; Cl" renal clearance.
* Significantly different from enoxacin alone (P < 0.05).
:I
i
.!!
:.0
>.~
.c'O
"0
~ a
t:
~~0
0.:;
cb\(
1
o~
.;
<
o~~--~--~~--~--~--~~--~
o 20 40 60 80
Absorbability of model drugs
(% absorbed in 1 h)
Fig. 5. Plot of the effect of bile salts on drug absorption against the absorbability of
the model drugs. (Adapted from KIMURA et al. 1985). Intestinal absorption of drugs
was examined in the presence of 20mM bile salts or sodium lauryl sulfate. Each
point represents the mean of at least three experiments. Drugs: 1, sulfaguanidine;
2, sulfanilamide; 3, sulfadimthoxine; 4, sulfanilacetamide; 5, sulfisoxazole; 6,
sulfapyridine; 7, sulfamethoxypyridazine; 8, sulfaphenazole; 9, sulfisomidine; 10,
sulfamethizole; 11, phenol red; 12, imipramine; 13, quinine; 14, 2-allyloxy-4-chloro-N-
(2-diethylaminoethyl)-benzamide; 15, NI,NI-anhydro-bis-(fJ-hydroxy-ethyl)biguanide;
16, metoclopramide; 17, procainamide. Filled circles, with sodium taurocholate; open
circles, with sodium cholate; filled triangles, with sodium taurodeoxycholate; open
triangles, with sodium lauryl sulfate
I. Passive Absorption
Various compounds with proposed enhancing effects on passive transcellular
as well as paracellular absorption have been investigated over recent years,
including oleic acid, sodium lauryl sulfate and bile salts (MURANASHI 1990).
KIMURA et al. (1985) summarized in a detailed report the effects of di- and
trihydroxy bile salts on the in situ absorption of a broad range of compounds
as depicted in Fig. 5. They proposed different mechanisms with regard to both
the observed enhancing as well as inhibitory effects of bile salts on drug
absorption. Sodium taurodeoxycholic acid (STDC) interacts most likely di-
rectly with the intestinal membrane by forming mixed aggregates with the
phospholipids in the lipid bilayer, therefore altering the membrane structure.
Drug Interactions in the Gastrointestinal Tract 29
0>-
f-
..... «
-1Q.
..... 0
CDO
« I 4 T
W-1
~
a:l.L.
WO
1
Q.
T
2 1
_T
1 T
.!. T
1 I
.J..
o
o 10 25 50 75 100
CONCENTRA nON LEUCINE (mM)
Fig. 7. Plot of the effect of the inhibitor L-Ieucine on the wall permeability of L-dopa.
The concentration of L-dopa remained constant at 0.1 mM whereas the concentration
of L-Ieucine varied from 0 to 100mM. (Adapted from SINKO et al. 1987)
Condition
Control
Vinblastine 0.5 mM
on APside ***
Vinblastine 0.5 mM
on BL side ***
Celiprolol 5 mM
on APside
Celiprolol 5 mM
on BL side
a 25 50 75 100 125
Basal-to-apical [3H)-vinblastine transport (%)
PV FA PV FA
Patient N
0 0 0 0 0
30 56 37 12 16
60 124 92 42 32
Pateint B
0 0 0 0 0
40 14 29 0 0
120 22 18 23 10
naringin produced much less of an effect in this study (Fig. 9) (BAILEY et al.
1993). AUC ratios between dehydrofelodipine (the main metabolite of
felodipine) and felodipine were reduced, suggesting inhibition of presystemic
metabolism. The relative contribution of intestinal metabolism in contrast to
hepatic metabolism remains to be determined for felodipine.
For cyclosporine (DUCHARME et al. 1995) and midazolam (KUPFERSCHMIDT
et al. 1995) on the other hand, it was demonstrated that grapefruit juice had no
effect on pharmacokinetic parameters such as AUC, systemic clearance and
elimination half-life when the drugs were administered intravenously. How-
ever, after oral administration of cyclosporine as well as midazolam together
with grapefruit juice an increase in bioavailability of 50% and 30%, respec-
tively, has been observed. The lack of effects on systemic clearance after
intravenous dosing suggests that oral bioavailability is improved by either
increased absorption or inhibition of gut-wall first-pass metabolism, with the
latter being more likely considering previous reports (KOLARS et al. 1991,
1992). However, one cannot completely rule out the effect on gastrointestinal
transit and increased solubilization due to bilary secretions as also being
partial explanations of these observations.
Clinically significant and severe interactions between azole antifungal
drugs and terfenadine, both substrates for CYP3A, have been reported
(HONIG et al. 1993; POHJOLA-SINTONEN et al. 1993). The decreased metabolism
of terfenadine to its active acid metabolite in the presence of either
itraconazole or ketoconazole results in increased plasma levels of
unmetabolized terfenadine. Terfenadine itself prolongs the QT interval and
increases the risk of torsade de pointes ventricular tachycardia. This metabolic
interaction might already occur on the intestinal level, but again the percent-
36 E. LIPKA et al.
15
-
0 0 WATER
:::;- • • GRAPEFRUIT JUICE
(5 )( )( NARINGIN SOL 'N
E
.s 10
0
Z
0
0
w
ii!: 5
9::
0
0
-'
W
IL
0
0.0 2.0 4.0 6.0 8.0
:::;- 20
~
E
.s
0 15
Z
0
0
w
~ 10
11.
0
0
-'
W
IL 5
0
a::
0
>
150
2.0 4.0 6.0 8.0
HOURS
Fig. 9. Plasma felodipine and dehydrofelodipine concentration-time profiles for the
three treatment groups receiving 5 mg felodipine with 200ml water, grapefruit juice or
naringin solution. Bars represent SEMs. Comparisons were made between results with
water and the two other treatments; *p < 0.025; **p < 0.01. (Adapted from BAILEY
et al. 1993)
References
Adjepon-Yamoah KK, Scott DB, Prescott LF (1973) Impaired absorption and
metabolism of oral lignocaine in patients undergoing laparoscopy. Br J Anaesth
45:143-147
Albert KS, Ayres JW, Disanto AR, Weidler DJ, Sakmar E, Halmark MR, StU RG,
DeSante KA, Wagner JG (1978a) Influence of kaolin-pectin suspension of digoxin
bioavailability. J Pharm Sci 67:1582-1585
Albert KS, DeSante KA, Welch RD, DiSanto AR (1978b) Pharmacokinetic evaluation
of a drug interaction between kaolin, pectin and clindamycin. J Pharm Sci
67:1579-1582
Algeri S, Cerletti C, Curcio M, Morselli PL, Bonollo L, Guniva G, Minazzi M, Minoli
G (1976) Effect of anticholinergic drugs on gastro-intestinal absorption of L-dopa
in rats and in man. Eur J PharmacoI35:293-299
Ambre JJ, Fischer U (1972) Effect of coadministration of aluminum and magnesium
hydroxides on absorption of anticoagulants in man. Clin Pharmacol Ther
14(2):231-237
Amidon GL, Lennernas H, Shah VP, Crison JR (1995) A theoretical basis for a
biopharmaceutics drug classification: the correlation of in-vitro drug product dis-
solution and in-vivo bioavailability. Pharmacol Res 12(3):413-420
38 E. LIPKA et al.
Ducharme MP, Warbasse LH, Edwards DJ (1995) Disposition of intravenous and oral
cyclosporine after administration with grapefruit juice. Clin Pharmacol Ther
57(5):485-491
Faloon WW, Chodos RB (1969) Vitamin B12 absorption studies using
colchicine, neomycin and continuous 57CoB 12 administration. Gastoenterology
56:1251
Fann WE, Davis JM, Janowsky DS, Sekerke HJ, Schmidt DM (1973) Chlorpromazine:
effects of antacids on its gastrointestinal absorption. J Clin Pharmacol13:388-390
Feldman S, Carlstedt BC (1974) Effect of antacid on absorption of enteric-coated
aspirin. JAMA 227:660-661
Fermaglich J, O'Doherty SS (1972) Effect of gastric motility on levodopa. Dis Nervous
Syst 33:624-625
Fleisher D (1995) Gastrointestinal transport of peptides: experimental systems. In:
Taylor MD, Amidon GL (eds) Peptide-based drug design. American Chemical
Society, Washington DC, pp 501-525
Forrest FM, Forrest IS, Serra MT (1970) Modification of chlorpromazine metabolism
by some other drugs frequently administered to psychiatric patients. BioI Psychiatr
2:53-58
Gibbons DO, Lant AF (1975) Effects of intravenous and oral propantheline and
metoclopramide on ethanol absorption. Clin Pharmacol Ther 17(5):578-584
Gomez DY, Wacher VJ, Tomlanovich SJ, Hebert MF, Benet LZ (1995) Clin
Pharmacol Ther (in press)
Gothoni G, Neuvonen PJ, Mattila M, Hackman R (1972) Iron tetracycline interactions:
effect of time interval between the drugs. Acta Med Scand 191:409-411
Grasela TH, Schentag JJ, Sedman AJ, Wilton JH, Thomas DJ, Schulz RW, Lebsack
ME, Kinkel A W (1989) Inhibition of enoxacin absorption by antacids or
ranitidine. Antimicrob Agents Chemother 33(5):615-617
Greenblatt DJ, Allen MD, MacLaughlin DS, Harmatz JS, Shader RI (1978) Diazepam
absorption: effect of antacids and food. Clin Pharmacol Ther 24:600-609
Greenspan R, MacLean H, Milzer A, Necheles H (1951) Antacids and aureomycin.
Am J Dig Dis 18:35037
Harcourt RS, Hamburger M (1975) The effect of magnesium sulfate in lowering
tetracycline blood levels. J Lab Clin Med 50:464-468
Hayes SL, Pablo G, Radomski T, Palmer RF (1977) Ethanol and oral diazepam
absorption. N Engl J Med 296:186-189
Heinrich HC, Oppitz KH, Gabbe EE (1974) Hemmung der Eisenabsorption beim
Menschen durch Tetracyclin. Klin Wochenschr 52:493-498
Ho NFH, Merkle HP, Higuchi WI (1983) Quantitative, mechanistic and physiologically
realistic approach to the biopharmaceutical design of oral drug delivery systems.
Drug Dev Ind Pharm 9(7):1111-1184
Hoffman WS, Dyniewicz HA (1945) The effect of alumina gel upon the absorption of
vitamin A from the intestinal tract. Gastoenterology 5:512-522
Honig PK, Wortham DC, Zamani K, Conner DP, Mullin JC, Cantilena LR (1993)
Terfenadine-ketoconazole interaction. Pharmacokinetic and electrocardiographic
consequences. JAMA 269(12):1513-1518
Hsing S, Gatmaitan Z, Arias 1M (1992) The function of Gp 170, the multidrug-
resistance gene product, in the brush border of rat intestinal mucosa. Gastroenter-
ology 102:879-885
Hu M, Amidon GL (1988) Passive and carrier-mediated intestinal absorption compo-
nents of captopril. J Pharm Sci 77(12):1007-1011
Hunter J, Hirst BH, Simmons NL (1993) Drug absorption limited by P-glycoprotein-
mediated secretory drug transport in human intestinal epithelial Caco-2 cell layers.
Pharmacol Res 10(5):743-749
Hurwitz A (1971) The effects of antacids on gastrointestinal drug absorption. II. Effect
on sulfadiazine and quinine. J Pharmacol Exp Ther 179(3):485-489
Hurwitz A, Schlozman DL (1974) Effects of antacids on gastrointestinal absorption of
isoniazid in rat and man. Am Rev Resp Dis 109:41-47
40 E. LIPKA et al.
lIett KF, Tee LBG, Reeves PT, Minchin RF (1990) Metabolism of drugs and other
xenobiotics in the gut lumen and wall. Pharmacol Ther 46:67-93
Ince P, Appleton DR, Finney KJ, Moorghen M, Sunter JP, Watson AJ (1988)
Verapamil sensitises normal and neoplastic rodent intestinal tissue to the
stathmokinetic effect of vincristine in vivo. Br J Cancer 57:348-352
Iseki K, lemura A, Sato H, Sunada K, Miyazaki K, Arita T (1984) Intestinal absorption
of several f3-lactam antibiotics. V. Effect of amino f3-lactam analogues and
dipeptides on the absorption of amino f3-lactam antibiotics. J Pharm Dyn 7:768-
775
Johnson BF, O'Grady J, Bye C (1978) The influence of digoxin particle size on absorp-
tion of digoxin and the effect of propantheline and metoclopramide. Br J Clin
Pharmacol 5:465-467
Karlson J, Kuo SM, Ziemniak J, Artursson P (1993) Transport of celiprolol across
human intestinal epithelial (Caco-2) cells: mediation of secretion by multiple
transporters including P-glycoprotein. Br J Pharmacol110(3):1009-1016
Kauffman RE, Azarnoff DL (1973) Effect of colestiopol on gastrointestinal absorption
of chlorothiazide in man. Clin Pharmacol Ther 14:886-889
Kimura H, Inui K-I, Sezaki H (1985) Differences in effects on drug absorption between
dihydroxy and trihydroxy bile salts. J Pharmacobio Dyn 8:578-585
Kolars JC, Awni WM, Merion RM, Watkins PB (1991) First-pass metabolism of
cyclosporin by the gut. Lancet 338:1488-1490
Kolars JC, Schmiedlin-Ren P, Schuetz JD, Fang C, Watkins PB (1992) Identification
of rifampicin-inducible P450IIIA4 (CYP3A4) in human small bowel enterocytes.
J Clin Invest 90:1871-1878
Koster AS, Frankhiujzen-Sierevogel AC, Noordhoek J (1985a) Glucuronidation of
morphine and six beta 2-sympathomimetics in isolated rat intestinal epithelial
cells. Drug Metabol. Dispos. 13:232-238
Koster AS, Hofman GA, Frankhiujzen-Sierevogel AC, Noordhoek J (1985b)
Presystemic and systemic intestinal metabolism of fenoterol in the conscious rat.
Drug Metabol Dispos 13:464-470
Krishnamoorthy R, Mitra A (1995) Peptide metabolism by gastric, pancreatic, and
lysosomal proteinases. In: Taylor MD, Amidon GL (eds) Peptide-based drug
design. American Chemical Society, Washington DC, pp 501-525
Kuhlmann J, Zilly W, Wilke J (1981) Effects of cytostatic drugs on plasma level and
renal excretion of beta-acetyldigoxin. Clin Pharmacol Ther 30:518-527
Kuo SM, Whitby BR, Artursson P, Ziemniak JA (1994) The contribution of intestinal
secretion to the dose-dependent absorption of celiprolol. Pharmcol Res 11(5):648-
653
Kupferschmidt HHT, Ha HR, Ziegler WH, Meier PJ, Kraehenbuehl S (1995) Interac-
tion between grapefruit juice and midazolam in humans. Clin Pharmacol Ther
58(1):20-28
Lavigne J-G, Marchand C (1973) Inhibition of gastointestinal absorption of p-
aminosalicylate (PAS) in rats and humans by diphenhydramine. Clin Pharmacol
Ther 14:404-411
Lebsack ME, Nix D, Ryeerson B, Toothaker RD, Welage L, Norman AM, Schentag JJ,
Sedman AJ (1992) Effects of gastric acidity on enoxacin absorption. Clin
Pharmacol Ther 52(3):252-256
Lennernas H (1995) Does fluid flow across the intestinal mucosa affect quantitative
oral drug absorption? Is there time for a reevaluation? Pharmacol Res 12
Lennernas H, Regardh CG (1993) Dose-dependent intestinal absorption and signifi-
cant intestinal secretion (exsorption) of the beta-blocker pafenolol in the rat.
Pharmacol Res 10(5):727-731
Lennernas H, Nilsson D, Aquilonius SM, Ahrenstedt 0, Knutson L, Paalzow LK
(1993) The effects of L-Ieucine on the absorption of L-dopa, studied by regional
jejunal perfusion in man. Br J Clin Pharmacol 35(3):243-250
Levy G, Rao BK (1972) Enhanced intestinal absorption of riboflavin from sodium
alginate solution in man. J Pharm Sci 61:279-280
Drug Interactions in the Gastrointestinal Tract 41
Peterson OL, Finland M, Ballou AN (1942) The effect of food and alkali on the
absorption and excretion of sulfonamide drugs after oral and duodenal adminis-
tration. Am J Med Sci 204:581-588
Pohjola-Sintonen S, Viitasalo M, Toivonen L, Neuvonen P (1993) Itraconazole pre-
vents terfenadine metabolism and increases risk of torsade de pointes ventricular
tachycardia. Eur J Clin PharmacoI45(2):191-193
Renwick AG (1982) First-pass metabolism within the lumen of the gastrointestinal
tract. In: George et al (eds) Clinical pharmacology and therapeutics: pre systemic
drug elimination. Butterworth, London, pp 3-28
Resetarits D, Bates TR (1979) Apparent dose-dependent absorption of chlorothiazide
in dogs. J Pharmacol Biopharmacol 7(5):463-470
Rietbrock N, Kuhlmann J, Leopold G (1983) Factors influencing the enteral absorption
of drugs. Inn Med 10:112-120
Rivera-Calimlim L, Dujovne CA, Morgan JP, Lasagna L, Bianchine JR (1970a) L-dopa
absorption and metabolism by the human stomach. Pharmakologist 12:269
Rivera-Calimlim L, Dujovne CA, Morgan JP, Lasagna L, Bianchine JR (1970b) L-
dopa treatment failure: explanation and correction. Br Med J 4:93
Rivera-Calimlim L, Morgan JP, Dujovne CA, Bianchine JR, Lasagna L (1970c) L-dopa
absorption and metabolism by the human stomach. J Clin Invest 49:79a
Rivera-Calimlim L, Dujovne CA, Morgan JP, Lasagna L, Bianchine JR (1971) Absorp-
tion and metabolism of L-dopa by the human stomach. Eur J Clin Invest 1:313-
320
Rivera-Calimlim L, Castaneda L, Lasagna L (1973) Effects of mode of management on
plasma chlorpromazine in psychiatric patients. Clin Pharmacol Ther 14:978-985
Robinson DS, Benjamin DM, McCormack JJ (1970) Interaction of warfarin and
nonsystemic gastrointestinal drugs. Clin Pharmacol Ther 12(3):491-495
Schreiner J, Altemeier WA (1962) Experimental study of factors inhibiting absorption
and effective therapeutic levels of declomycin. Surg Gynecol Obstet 114:9-14
Schuna A, Osman MA, Patel RB, Welling PG, Sundstrom WP (1983) Influence of food
on the bioavailability of penicillamine. J Rheumatol 10:95-97
Seed JC, Wilson CE (1950) The effect of aluminum hydroxide on serum aureomycin
concentrations after simultaneous oral administration. Bull Johns Hopkins Hosp
86:415-418
Seneca H, Peer P (1965) Enhancement of blood and urine tetracycline levels with a
chymotrypsin-tetracycline preparation. J Am Geriatr Soc 13:708-717
Sinko PJ, Hu M, Amidon GL (1987) Carrier mediated transport of amino acids, small
peptides, and their drug analogs. J Contr ReI 6:115-121
Sorby DL, Liu G (1966) Effects of adsorbents on drug absorption. II. Effect of an
antidiarrhea mixture on promazine absorption. J Pharm Sci 55:504-510
Tam YK (1993) Individual variation in first-pass metabolism. Clin Pharmacokinet
25(4):300-328
Tsuji A (1995) Intestinal absorption of j3-lactam antibiotics. In: Taylor MD, Amidon
G L (eds) Peptide-based drug design. American Chemical Society, Washington
DC, pp 501-525
Tyrer JH, Eadie MJ, Sutherland JM, Hooper WD (1970) Outbreak of anticonvulsant
intoxication in an Australian city. Br Med J 4:271-273
Veno K, Tanaka K, Tsujimura K, Morishima Y, Iwashige H, Yamazaki K, Nakata I
(1993) Impairment of cefdinir absorption by iron ion. Clin Pharmacol Ther
54(5):473-475
Wacher VJ, Wu CY, Benet LZ (1995) Mol Carcinog (submitted)
Wagner JG (1969) Aspects of pharmacokinetics and biopharmaceutics in relation to
drug activity. Am J Pharm 141:5-20
Waisbren BA, Hueckel JS (1950) Reduced absorption of aureomycin caused by alu-
minium hydroxide gel (Amphojel). Proc Soc Exp BioI Med 73:73-74
Walter E, Kissel T, Amidon GL The intestinal peptide carrier: a potential transport
system for small peptide derivated drugs. Adv Drug Deliv Rev (accepted)
Drug Interactions in the Gastrointestinal Tract 43
Watkins PB, Wrighton SA, Schuetz EG, Molowa DT, Guzelian PS (1987) Identifica-
tion of glucocorticoid-inducible cytochromes P-450 in the intestinal mucosa of rats
and man. J Clin Invest 80:1029-1036
Weinberg ED (1957) The mutual effects of antimicrobial compounds and metallic
cations. Bacteriol Rev 21:46-68
Welling PG (1984) Interactions affecting drug absorption. Clin Pharmacol 9:404-434
Wetterich U, Mutschler E, Spahn-Langguth H, Langguth P (1995) Evidence for intes-
tinal secretion of the f3-adrenoceptor antagonist talinolol: data from humans and
studies with Caco-2 cells. Naunyn Schmiedenbergs Arch Pharmacol 351
[Suppl]:R1
Whitehouse LW, Paul CJ, Coldwell BB (1975) Effects of ethanol on diazepam distribu-
tion in rats. Res Commun Chern Pathol Pharmacol12:221-242
Williamson RM, Oxender DL (1995) Molecular biology of amino acid, peptide and
oligopeptide transport. In: Taylor MD, Amidon GL (eds) Peptide-based drug
design. American Chemical Society, Washington DC, pp 501-525
Yamaguchi T, Ikeda C, Sekine Y (1986a) Intestinal absorption of a f3-adrenergic
blocking agent nadolol. I. Comparison of absorption behavior of nadolol with
those of other f3-blocking agents in rats. Chern Pharm Bull 34(8):3362-3369
Yamaguchi T, Ikeda C, Sekine Y (1986b) Intestinal absorption of a f3-adrenergic
blocking agent nadolol. II. Mechanism of the inhibitory effect on the intestinal
absorption of nadolol by sodium cholate in rats. Chern Pharm Bull 34(9):3836-
3843
Yamaguchi T, Ikeda C, Sekine Y (1986c) Intestinal absorption of a f3-adrenergic
blocking agent nadolol. III. Nuclear magnetic resonance spectroscopic study on
nadolol-sodium cholate micellar complex and intestinal absorption of nadolol
derivatives in rats. Chern Pharm Bull 34(10):4259-4264
Yee S, Amidon GL (1995) Oral absorption of angiotensin-converting enzyme inhibi-
tors and peptide prodrugs. In: Taylor MD, Amidon GL (eds) Peptide-based drug
design. American Chemical Society, Washington DC, pp 501-525
CHAPTER 3
Drug-Food Interactions
Affecting Drug Absorption
P.G. WELLING
A. Introduction
The intent of this book is to present current information on drug interactions
influencing drug absorption, distribution, elimination, and activity. Consistent
with this, the original plan of the authors concerned was to present a single
chapter on interactions affecting drug absorption which would include drug-
drug interactions and also drug-food interactions. After examining the quan-
tity of material that has been published on these two closely related but
distinct topics, it was agreed that drug-drug interactions and drug-food inter-
actions affecting drug absorption would be better presented in two separate
chapters.
This chapter then, is devoted exclusively to drug-food interactions affect-
ing drug absorption. No attempt has been made to address the related topic of
the effect of drugs on food and nutrient absorption. Material on this topic has
been reviewed elsewhere (BASU 1988; ROE 1989; TROVATO et al. 1991).
While a number of studies on drug-food interactions were reported
earlier, the subject did not achieve full recognition in scientific and regulatory
circles until the first major review on this topic (WELLING 1977). Since that
time the number of studies examining various aspects of drug-food interac-
tions affecting drug absorption has increased dramatically and many of the
results obtained from these studies have been quite spectacular, not only in the
extent of some interactions but also in their unexpected and totally unpredict-
able nature. The topic has been reviewed extensively, not only in terms of
interactions of particular drugs or families of drugs (WELLING 1989, 1993;
PFEIFER 1993; SORGEL and KINZIG 1993; WELLING and TSE 1982; WILLIAMS et al.
1993), but also relating to clinical consequences and the management of
drug-food interactions in the clinical environment (LASSWELL and LORECK
1992; NEUVONEN and KMSTO 1989). The number of reviews on this topic
reflects increased awareness in scientific, regulatory, and clinical circles of its
importance.
The original goal of many investigators in this area, including the writer,
was to establish mechanisms involved in drug-food interactions and also to use
data obtained from a variety of situations to establish rules or guidelines to
predict the nature and extent of interactions in terms of circulating drug or
metabolites. As with so many scientific goals this has proven to be elusive.
46 P.G. WELLING
After almost 20 years of research it is still difficult, and many times impossible,
to predict the nature of drug-food interactions and the literature is replete with
surprises, and unpredictable results.
Based on current knowledge, the unpredictable nature of various inter-
actions is probably related, not only to the complex environment in which
interactions occur, but also to the many different ways in which they have been
examined. Some of them are summarized here.
I. Food
The type and size of meal may have a marked effect on the nature of a drug-
food interaction. One might not expect that a meal prepared in England, in
India, or in Japan would act similarly, nor might a breakfast compared to an
evening meal. Similarly liquid meals, which are so often used in an attempt to
obtain mechanistic information, might have a totally different effect on drug
absorption than solid meals, which are usually more clinically relevant. Also
the time interval between eating and medication will influence the nature and
extent of a drug-food interaction.
moved from the stomach into the intestine mainly during phase 3. Depending
on when a solid meal is ingested relative to the MMe, the gastric residence
time may vary from a few minutes to 2-3 h (EWE et al. 1989). Ingested food
changes gastric motility to a postprandial pattern during which time gastric
residence time is increased. Gastric residence time is increased by large, par-
ticularly solid meals and by chyme of low pH, high osmolality, and high fat
content. Residence time is also influenced by hot meals.
While solid foods tend to delay gastric emptying, non-nutrient liquid
meals, for example, a fluid volume ingested with medication, may have the
opposite effect. As described previously in the review by WELLING (1977), the
stomach appears to empty liquid meals into the duodenum in apparent first-
order fashion. Distension of the stomach is the only natural stimulus known to
increase stomach emptying and the observed faster emptying rate with in-
creasing fluid volume can be rationalized in terms of varying tension at recep-
tors in the stomach wall (HOPKINS 1966). The presence of ingested food also
promotes gastric secretion of hydrochloric acid. Once food passes from the
48 P.G. WELLING
stomach into the small intestine it has a stimulating effect on intestinal motil-
ity, on digestive enzyme secretion, and also, particularly in the case of fatty
meals, on bile secretion.
The influences of altered gastric and intestinal motility, the one decreased
and the other increased, and increasing GI secretions with ingested food,
might be expected to have a number of possible effects on drug absorption. All
of these have been proposed, in one way or another, to explain observed
results of drug-food interactions. Delayed gastric emptying will likely delay
absorption of drugs that are absorbed predominantly from the small intestine,
but not of drugs that are efficiently absorbed from the stomach. It may delay
the absorption of acidic compounds or drugs in enteric-coated formulations by
delaying drug transit from the acidic gastric contents to the relatively alkaline
region of the small intestine. On the other hand, delayed gastric emptying
might increase systemic availability of compounds that are slowly soluble at
acidic gastric pH by permitting more material to dissolve in the stomach
before passing into the small intestine. Compounds that are unstable in acidic
pH are likely to be degraded as a result of prolonged residence in the stomach
regardless of the extent to which pH may be lowered, or raised, by the intake
of food. The latter point is important because, while solid foods stimulate
acidic secretion, the buffering effect of different types of meals may minimize
any change in pH and could even give rise to a transient increase in gastric pH.
Increased intestinal motility in the fed state may have the effect of increas-
ing dissolution of solid particles and could also decrease the mean diffusional
path of dissolved molecules to the intestinal mucosa. On the other hand, it
could increase the transit rate of molecules from the efficient absorption
region of the small intestine to the less efficient region of the large intestine,
thus negatively impacting absorption efficiency.
The effect of fluid volume on GI physiology adds another dimension to the
complexity of drug-food interactions. It has already been noted that large
volumes of non-nutrient fluids empty from the stomach at a faster rate than
small volumes. The large volume itself tends to accelerate dissolution and also
to accelerate transfer of both dissolved and undissolved drug into the small
intestine. The presence of large fluid volumes in the intestine may continue to
increase drug absorption by providing a more liquid environment and also by
a solvent drag effect from the serosal to the mucosal side of the intestinal
membrane. On the other hand, and from a physicochemical perspective, a
large fluid volume might be expected to delay or reduce absorption due to a
reduced serosal-mucosal drug concentration gradient. In the original review
on this subject (WELLING 1977) it was noted that the positive effects of large
fluid volumes appear often to outweigh the negative effects, and more
examples of increased drug absorption when they are administered with large
fluid volumes have appeared in the literature to support this argument. The
role of gastric motility in drug absorption, with particular reference to the
definition of "fasting" and "non-fasting" drug administration, has recently
been reviewed by WALTER-SACK (1992).
Drug-Food Interactions Affecting Drug Absorption 49
Table 2. Direct drug-food interactions that may influence drug absorption. (From
WELLING 1993)
more protein-derived drugs, direct competition for active carriers may occur
between food protein and protein fragments and drug molecules, inevitably
giving rise to decreased drug systemic availability.
Given the multifactorial nature of drug-food interactions, and the large
number of variables that need to be addressed in one way or another in studies
of this type, it is not surprising that variable and often conflicting results are
obtained from different laboratories. In order to better understand and inter-
pret these types of studies it is essential that study conditions be carefully
controlled and completely described. The writer makes no plea for uniformity
in studies, on the contrary. Only by studying interactions under widely varying
conditions will it be possible to completely understand them, together with
their mechanistic and clinical implications. Nonetheless, complete study de-
tails must be reported just as in the recording of a chemical or physical
experiment. Different studies have different goals. For example administering
a drug together with the well-known English breakfast addresses a clear
clinical question, relevant at least to those who indulge in extravagances, but
does not attempt to address the mechanistic question. Administering a drug
together with a high-protein fixed-volume liquid meal, on the other hand,
attempts to address the mechanistic question but tends to be clinically irrel-
evant. Which study is the most important depends on one's perspective. How-
ever, both study types are necessary in order to completely understand the
phenomenon. It is interesting to note that the extent to which drug absorption
may be altered by the presence of food, while in some cases may be slight or
even negligible, may in other cases be far greater than would be permitted by
any regulatory agency if it were considering interchangeability of generic
products. The problem of drug-food interactions has been exacerbated by the
introduction of controlled release oral products. These devices may serve a
useful clinical role in generating desirable plasma drug profiles, possibly lead-
ing to better control of pathological conditions, and also increasing patient
compliance. However, they also present a greater quantity of drug to the
patient per single dosing unit than conventional dosage forms, and are de-
signed to deliver drug at a controlled rate over a prolonged period. As these
dosage forms are also given less frequently than conventional formulations, a
Drug-Food Interactions Affecting Drug Absorption 51
SK & F 106203 Tablet 150mg SO 12 healthy male Plasma levels reduced NICHOLS et at.
subjects and delayed (1992)
Sotalol 160mg SO 18 healthy subjects Standard meal 36h plasma Bioavailability reduced HANYOK (1993)
approximately 20%
Sulpiride Film-coated 100mg SO 3 healthy male Standard light, 100ml water With meal 48 h, urine Cumulative urinary SHINKUMA et al.
tablet, solution subjects medium and heavy excretion reduced by 30% (1990)
mealsd following both dosage
forms
Tacrine Capsule 40mg SO 24 healthy subjects Standard breakfastd 8 fluid 15 min and 2h 14 h plasma Cmax reduced 45% and WELTY et al.
ounces water after starting meal AUe 23% when taken (1994)
15 min after starting meal.
Cmax reduced 35 % and
AU C 21 % when taken
2 h after starting meal
Tetracycline Capsule 250mg SO 9 healthy subjects Standard meal and 200mi water Immediately after 72 h urine Urine excretion reduced COOK et a1.
high-calcium meal 40% by standard meal and (1993)
Mexican meald 55% by Mexican meal
Verapamil SR tablet 240mg SO 9 halthy male Immediately after 30h serum 48 % decrease in C max and HOON et al.
subjects meal 30% decrease in AUe, (1992)
also altered electro
cardiographic effects
Zidovudine 100mg, SO 8 male 2500 kJ breakfast, 200ml water Mid-meal 6h plasma Cma~ and AUe reduced LOITERER et al
250mg AIDS patients fat content 40 g 64% and 29%, (1991)
respectively
Zidovudine Capsule 250mg SO 13 AIDS patients, High-fat liquid meal 600ml water Immediately 4 h plasma C max reduced 50%, UNADKAT et al.
IOM,3F given as milk shaked after meal and urine T max increased (1990)
approximately threefold
Zidovudine Capsule 100 mg, SO 25 patients, 24M 3F, Standard breakfastd 100ml water Immediately after 6 h plasma, C max and AUe reduced RUHNKE et al
250mg with HIV infection meal 24 h urine 35% at 100-mg dose, (1993)
Cmax reduced 73% and
AUC 14% at 250-mg dose
Zidovudine 200mg SO HIV-I-positive After meal 4h serum Serum C max reduced UEDA et aL
patients 50% and AUC 38%. (1993)
Urinary recovery reduced
by 19%. Serum levels of
glucuronide also reduced
but urinary recovery
unchanged
882C 200mg SO 15 healthy male Standard breakfast em.. reduced 16%, AUC PECK et aL
subjects reduced 10%. Food (1993)
reduced variability in
AUC
15
E *
-1
~
E
*
1/ -l. . j
~
"'C
.;:
0
10
::c<.I
E
:s
'c0
c
~
..Q 5
E
« I
I
E I
...
:s I
~
til
I
I
0
0 60 120 180
Time (min)
Fig. 1. Mean serum versus time concentrations (±SE) of ambenonium chloride after
oral administration of lOmg ambenonium chloride to six myasthenia gravis patients
underfasting (0) and nonfasting (e) conditions. *P, <0.05; **P, <0.01. (From OHTSUBO
et al. 1992)
Significant differences were observed between fed and fasted serum concen-
trations at 1.5, 2, and 3 h (P < 0.05) following the lO-mg dose. Following the
5-mg dose, significant differences were observed at 1 (P < 0.005),1.5,2, and 3 h
(P < 0.05).
Ambenonium chloride is a quaternary ammonium compound and is un-
likely to be well absorbed, even in the fasting state. Further reduction of
absorption due to food could be due to a number of factors, but these were not
examined in this study. Despite the rather dramatic food effect, relationships
between serum drug levels and relative clinical effect (changes in muscle
strength) were variable and inconclusive. It is nonetheless recommended that
ambenonium chloride dosage be adjusted during non-fasting treatments when
the adjusting the optimum regiment for patents with myasthenia gravis.
Previous studies with atenolol have shown that absorption is reduced by
food (MELANDER et al. 1979). In this respect the very hydrophilic atenolol
molecule differs from the lipophilic compounds of this class, metroprolol and
propranolol, both of which undergo extensive pre systemic metabolism and
whose absorption is increased by food. The mechanism by which food de-
creases atenolol absorption was addressed in a study conducted in healthy
volunteers who received single 50-mg doses of atenolol as a commercial tablet,
or as capsules containing bile acids (BARNWELL et al. 1993). In this study the
bile acid formula reduced mean atenolol plasma e max by 28% and AVe by
56 P.G. WELLING
15
~
5
QJ
~
.§ 10
::cu **
E
::::I
·2 *
l-L-t~
o
c:
~
E
5 *
<C
.
E
::::I
QJ
III I
/ -----~
I,A--~--~--~-- __~
O~~---,------~----~
o 60 120 180
Time (min)
Fig. 2. Mean serum versus time concentrations (±SE) of ambenonium chloride after
oral administration of 5 mg ambenonium chloride to seven myasthenia gravis patients
under fasting (0) and nonfasting (.) conditions. *P, <0.05; **P, <0.01. (From OHTSUBO
et al. 1992)
a 22% reduction in mean Cmax values and a 16% reduction in urinary excretion
of cefprozil. In the second study, simultaneous administration of an elemental
diet Enterued, composed of oligopeptide (egg white hydrolysate), an elemen-
tal diet Hepan ED composed of amino acids, and also a mineral solution
containing neither pep tides nor vitamins had a similar effect on ceftibuten.
Plasma level data in rats, and also in situ rat jejunal loop studies, confirmed the
effect of these test meals on ceftibuten absorption. These results contrasted
with those from cephalexin, which was unaffected by Enterued formula, show-
ing that the inhibitory effects of Enterued on absorption were not due to
inhibition of a peptide transport system.
Administration of a standard breakfast moderately reduced the systemic
availability of oral 2-chloro-2'-deoxyadenosine (CdA) in male patients with
leukemia (ALBERTIONI et al. 1993). The Cmax of CdA in plasma was reduced by
40% while the AVC was reduced by 9% by food. In this study, pretreatment
with omeprazole did not significantly influence CdA bioavailability, or
interindividual variability in the fasting state. The absolute bioavailability of
CdA is only 50% when administered to fasting patients with and without
omeprazole. CdA has a low pKa such that most of the drug will be ionized in
the gut, hindering absorption. The drug may also undergo first-pass clearance
in the gut wall and/or the liver. Reduced systemic availability in the presence
of food may be related to slower absorption, resulting in a greater proportion
of absorbed drug undergoing presystemic metabolism. Reduced absorption
due to food may be a possible explanation for reduced antiplatelet activity of
cicaprost (BELCH et al. 1993). Healthy volunteers received varying doses of
cicaprost at 9 a.m., 2 p.m., 7 p.m., and again at 9 a.m. the next day. Antiplatelet
activity was dose related but the effect was attenuated following the 2 p.m. and
7 p.m. doses, which were taken after heavier meals than the 9 a.m. doses. An
alternative explanation is that tachyphylaxis may occur during the second (2
p.m.) and a third (7 p.m.) dose, causing reduced efficacy, while the 14-h gap
between the 7 p.m. dose and the next 9 a.m. dose may have been sufficient for
platelet sensitivity to return to normal. More studies are necessary to differen-
tiate these possible mechanisms. Although neither a standard breakfast nor a
high-fat, high-calcium breakfast altered ciprofloxacin absorption (FROST et al.
1989), absorption was markedly impaired by co administered milk and yoghurt
(NEUVONEN et al. 1991). Systemic availability was reduced 35% by
co administered 30ml milk and 36% by 300ml yoghurt compared to 300ml
water. Ciprofloxacin together with other fluoroquinolones is known to bind to
heavy metals including calcium to form an insoluble chelate. The reason for
the different results from the two studies is claimed to be due to calcium in
milk and yoghurt being in liquid form compared to the previous study in which
most of the high calcium present was in solid form, for example, in cheese. This
suggests, that, for at least some dairy products, ciprofloxacin is affected to a
similar extent as tetracycline derivatives (MATTILA et al. 1972). Plasma levels of
ciprofloxacin taken with milk, yoghurt, and water are shown in Fig. 3.
58 P.G. WELLING
The timing of food ingestion relative to dosing had a marked effect on the
absorption of the purine nucleoside analogue didanosine (KNUPP et a1. 1993).
This study, conducted in ten men seropositive for human immunodeficiency
virus (HIV) but free of AIDS symptoms, examined the absorption of
didanosine following overnight fast, 13 min before, 1 h before, 1 h after, and 2 h
after a standard high-fat, high-calorie breakfast. There were no significant
differences among the fasting, 30min, and 1 h before meal doses. However,
Cmax , AUC(O-oo), and urinary excretion were significantly reduced following
the two postprandial doses. The mean profiles following the five treatments
are summarized on a logarithmic scale in Fig. 4.
Decreased bioavailability after postprandial administration may have
been due to prolonged gastric retention leading to increased degradation of
the acid-labile didanosine, possible interference with active transport, or direct
interaction of didanosine with one or more meal components. In a separate
study using a chewable tablet form of didanosine in male subjects seropositive
for HIV, the bioavailability of didanosine was reduced approximately twofold
when it was administered 5 min after a substantial, standardized breakfast
o~~~~--~--~------~----~------~--~
o 1 2 3 4 6 8 10 24
Time (hr)
Fig. 3. Mean plasma versus time concentrations (±SE) of ciprofioxacin in seven sub-
jects following a single 500-mg oral dose of ciprofioxacin with 300ml milk (e), yoghurt
(.), or water (0). (From NEUVONEN et a1. 1991)
Drug-Food Interactions Affecting Drug Absorption 59
2000,-------------------------------~
-0- Fasting
-0- 30 min Before
-b- 1 hr Before
...... 1 hr After
-(too 2 hr After
~5
Q,j
c
'iii
g 200
:a'"
"C
'~E"
c:::
2~--~----_+------~------~--~
o 2 4 6
Time (hr)
Fig. 4. Mean plasma versus time concentrations of didanosine in ten patients following
a single 300-mg oral dose of a chewable tablet under fasting conditions or at various
times relative to a high-fat, high-calorie meal. (From KNUPP et al. 1993)
(SHYU et al. 1991). The results obtained in these two studies have led to the
recommendation that didanosine be administered under fasting conditions.
A milder food effect was reported for the novel quinazoline anti-
hypertensive agent doxazosin. Following single I-mg doses with a standard-
ized light breakfast, mean emax in 12 hypertensive subjects was reduced 18%
and the AVe by 12% compared to fasting. The acute fall in blood pressure
following doxazosine doses was not significantly affected by food, but there
was a trend toward lower instances of complaints after this treatment. Apart
from that, the modest food effects observed in this study are unlikely to be
clinically significant. A more serious clinical effect with flecainide was possibly
attributed to a milk drug interaction (RUSSEL and MARTIN 1989). Toxicity in
the form of ventricular tachycardia occurred in a baby boy when dextrose was
substituted for milk foods during flecainide therapy. Serum flecainide levels,
based on single point determinations, were doubled from 990.ug/ml to 1824.ug/
60 P.G. WELLING
c
·0
ro
)(
0 0.4
...0
'=
c
ro
E
<II
ftI
0.2
'6:
0
0 1 2 3 4 6 8 10
~24
Time (hr)
Fig. S. Mean plasma versus time concentrations (±SE) of norfloxacin in seven subjects
following a single 200-mg oral dose of norfloxacin with 300ml milk (e), yoghurt (.), or
water (0). (From KIVISTO et al. 1992)
62 P.G. WELLING
al. 1993). Clearly both diets in this study contained sufficient free calcium to
markedly reduce systemic availability of tetracycline.
Contradictory effects were observed on pravastatin pharmacokinetics and
pharmacodynamics when the drug was taken with meals (PAN et al. 1993). In
a study conducted in 24 hypercholesterolemic men, mean Cm• x dropped by
49% and AUC by 31 % in the presence of food compared to the fasting state.
However, reduction in mean total cholesterol and low-density lipoproteins
was identical whether pravastatin was taken with or before food. The reduc-
tion in pravastatin bioavailability in these hypercholesterolemic patients was
similar to that observed in a preliminary study in healthy volunteers. The lack
of effect on blood lipids may have been due to food increasing extraction of
pravastatin by the liver, which is also the primary site of cholesterol synthesis,
LDLIcholesterol clearance, and also pravastatin activity. Other studies within
this category have reported 60% reduction in the already poorly absorbed
renin inhibitor remikiren (WEBER et al. 1993), a modest reduction in the
bioavailability of the leukotriene receptor antagonist SK&F 106203 (NICHOLS
et al. 1992), and moderately reduced plasma levels of rufloxacin (SEGRE et al.
1992) in the presence of food. In the last case, however, although mean Cm• x
and AUC values were decreased by 16% and 33%, respectively, urinary
excretion of unchanged drug increased from 16% to 26% of the administered
dose. The increase of urinary excretion suggests that lower plasma levels of
rufloxacin may not have been due to reduced drug absorption.
The beta-blocking agent sotalol appears to behave more like atenolol
(BARNWELL et al. 1993) than metoprolol or propranolol in that its systemic
bioavailability is reduced by approximately 20% by administered food. Like
atenolol, sotalol undergoes little or no first-pass metabolism so that any
change in hepatic blood flow due to food is not likely to have a positive effect
on systemic availability.
Formulation was shown to make a negligible contribution to the nature of
drug-food interaction for the neoleptic agent sulpiride (SHINKUMA et al. 1990).
Ingestion of a film-coated tablet or a solution with a standard meal resulted in
30% reduction in systemic availability. Further studies with the solution dos-
age form showed that the food effect increased with increasing meal size,
inhibition increasing from 21 % with a light meal, to 30% with a medium meal,
to 40% with a heavy meal. The systemic availability of the cognition agent
tacrine was reduced by approximately 20%, and peak plasma levels by 40%
when administered up to 2h following the start of a standard breakfast (WELTY
et al. 1994). However, administration with meals reduced gastrointestinal side
effects, thus improving patient tolerance. In the case of verapamil, food has
been shown not only to reduce systemic availability from a sustained release
formulation, but also to alter electrographic effects following a single drug
dose. There was no evidence of exaggerated release or "dose dumping" in this
study.
A number of studies have examined the effect of food on the absorption
and pharmacokinetics of the HIV agent zidovudine (LOITERER et al. 1991;
Drug-Food Interactions Affecting Drug Absorption 63
UNADKAT et al. 1990; PUHNKE et al. 1993; UEDA et al. 1993). All of these
studies, conducted in laboratories in Japan, Germany, and the United States,
have reported significantly delayed and reduced absorption with remarkable
consistency across studies. Typically, Tmax is increased from O.S to l.S-2.Oh,
while Cmax and AUC are decreased by approximately SO% when zidovudine is
administered with a meal compared to fasting. Variability in plasma levels is
also increased by food. While no direct relationship has been established
between circulating levels and efficacy of zidovudine, doses found to be effec-
tive in AIDS patients generally achieve peak serum levels of ~0.27 ,ug/ml.
However, lower and more prolonged levels, as observed when zidovudine is
taken with meals, may maintain efficacy while reducing toxic side effects
(UNADKAT et al. 1990).
For the last compound in this category, 882C (1-(j3-D-arabinofuranosyl)-S-
(1-propynyl)uracil), which is under investigation for treatment of varicella
zoster virus infections, food caused a significant reduction in Cmax and a modest
reduction in AUC in healthy volunteers (PECK et al. 1993). The effects ob-
served in this single-dose study led to the recommendation that effective
concentrations of this agent can be maintained without food-related
restrictions.
Acetorphan l00mg SOb 9 healthy subjects Standard meal, 20 min after meal - T min for enkephalinase BERGMANN
800kcal activity 180 min after food et al. (1992)
compared to 80 min for
acetorphan alone
Albuterol Volmaxand 4mg SO 23 healthy male Standard 250ml water Immediately after 30h plasma emu reduced 19% for HUSSEyet a1.
Proventil subjects breakfastc meal Volmax and 21 % for (1991)
Repetabs Proventil. Tmax and tlag
Albuterol sulfate delayed
5-Aminosalicylic Tablet SOOmg MDd 12 healthy male Breakfast, 100 ml water Immediately after 48h plasma Systemic absorption delayed DE MEyand
acid subjects afternoon snack, meals and urine but not reduced by food MEINEKE (1992)
evening snack,c
standardized
Aniracetam Tablet 1200mg SD emax reduced 50%, TIDaJI. RONCARl (1993)
extended from 0.6 to 1.1 h. HONMA et a1.
AVC of metabolites (1986)
unaffected by food
Beta- Lanirapid 0.2mg SO 9 healthy male Standard breakfast' 150ml water 30 min before or 120h serum, Absorption rate significantly TSUTSUMI et aI.
methyldigoxin subjects 560 kcal. Other after meal 240 h urine reduced after meal. Mean (1992)
meals also Cmax reduced 46%, Ave
controlled unaffected. V rinary
excretion slightly decreased
Cefaclor Capsules 500mg SD 8 healthy male Two standard 30 min after meal 6 h plasma IillIX
C reduced, Tmax OGUMA et a1.
subjects breakfasts. cOne and urine prolonged. Effect increased (1991)
based on rice, the with larger meal size. Rice
other based on eggs meal affected absorption
and bread. Six more than bread meal
months later
repeated, rice meal
reduced, bread
meal increased
Cefdinir 10% fine 3 mg/kg SD 8 children 30 min before or Cmax reduced 43%. Tmax MOTOHIRO et aI.
granules after meal increased from 2 h to 4-5 (1992)
h. AVC reduced by 15%.
Urinary recovery marginally
reduced
Cefdinir 5% fine granules 3 mg/kg SD 20 schoolchildren - 30 min before Cmax reduced 30%, Tmax NAKAMURA and
(14 children) or increased from 2 to 3.67 h. IWAI (1992)
30 min after V rinary recovery unchanged
(6 children) meal
Cefdinir 5% fine granules 3 mg/kg SD 25 younger 30 min before (19 - CroaK reduced 52%, Tmax NAKAMURA and
children children) or 30 increased from 2.1 to IWA! (1992)b
min after (6 3.3 h. Urinary recovery
children) meal reduced by 48 %
Cefdinir 5% fine granules 3 mg/kg SD 15 infants 30 min before (7 No significant differences NAKAMURA and
infants) or 30 min between groups IWA! (1992)
after (8 infants)
meal
Cefprozil Capsule 100mg SD 15 healthy male Standard breakfast' 200 ml water 30 min after meal 12h plasma, Tmax increased from 1.5 to SHUKLA et a1.
subjects 24h urine 2 h. Cm~' AUC, urinary (1992)
excretion not affected
Cefprozil Capsule 250mg SD 12 healthy male Standard breakfast C 150 ml water 15 min after meal 8h plasma Slight reduction in emu' BARBHAIYA
subjects mIDI.
Mean T increased from et a1. (1990)
1.2 to 2h
Cefaclor Capsule 250mg SD 12 healthy male Standard breakfast' 150ml water 15 min after meal 5 h plasma CmllJr reduced 51 %, Tmax BARBHAIYA et
subjects increased from 0.6 to 1.3 h a1. (1990)
CL 275838 Tablet 50mg SD 10 healthy male Standard breakfastC 150ml water 30 min after meal 48h plasma Tm~ delayed 2 h. Cm~' CONFALONIERI
subjects AUC unaffected. et a1. (1992)
Dic10fenac Capsule 50mg SD 8 healthy male Standard breakfast 150ml water Together with 10 h serum e max reduced 61 %, Tmu TERHAAG et a1.
subjects meal increased from 0.8 to (1991)
2.4h. AUC unaffected
Diclofenac Hydrogel bead 150mg SD 12 healthy male Standard breakfastC 200ml water 30 min after meal 24h plasma Cmax reduced 38 %, Tmax. THAKKER et al.
capsule subjects increased from 1.85 to (1992)
6.25 h. AUC unaffected
Diltiazem Pellets in capsule 240mg SD 8 healthy subjects Standard breakfast, 100ml water After meal 24h plasma Tmax delayed from 7.4 to WILDING et a1.
4M,4F lunch, dinner: 9.7 h. C_ and AUC (1991a)
unaffected
Doxycycline Pellets in capsule 100mg SD 20 healthy male Standard breakfast 180 ml water With breakfast 48 h plasma Lag time increased from WILLIAMS et al.
subjects and urine 0.55 to 1.1 h, Tmuincreased (1990)
from 1.8 to 3.1 h. C=" AUC,
and urinary excretion not
significantly affected
Erythromycin Enteric-coated 400mg b.i.d. SDandMD 14 healthy Standard light and Immediately after 12 h plasma After SD, both meals JARVINEN et a1.
acistrate tablet for 4 days subjects 4M, lOF heavy breakfastC meal delayed absorption. In seven (1992)
subjects no absorption
occurred in 12 h. After
MD food effect diminished
Fadrozole Tablet 12mg SD 9 healthy Standard breakfastC 240 ml water Within 20 min of 36 h plasma Cmax reduced by 15%, CHOI et al.
Caucasian completing meal T max increased from 1.8 to (1993)
SUbjects, 7M, 2F 2.5 h. AUC not affected
Table 4. Continued
Drug Dosage form Dose Dosage regimen Subjects Food details Fluid Time interval Sampling Result of food Reference
volume duration administration
Famotidine Tablet 40mg SD 6 p.m. or 10 healthy Supper given at With meal Variable Absorption delayed MARGALITH
9 p.m. subjects, 4M, 6F 6 p.m. or 9 p.m. plasma somewhat when meal and et aI. (1991)
drug coadministered at
6 p.m. No delay when meal
and drug coadministered at
9 p.m.
Flurbiprofen Tablet 100mg SD 12 healthy male 30 ml apple juice' 180ml water After the apple 48 h serum Gastric emptying time DRESSMAN
subjects juice almost doubled by meal. et al. (1992)
Slower initial absorption
followed by more rapid
second absorption phase
Fluvastatin Solution or 10mg SD 16 healthy male Standard high-fat Immediately 12 h blood e max reduced 69% to 74% SMITH et al.
capsule subjects breakfast after meal and TmaX increased more (1993)
than fourfold from solution.
Cmax reduced 50%--60%
and TmaX increased three-
to fourfold from capsule.
Bioavailability decreased by
15%-25%
Fluvastatin Capsule 20mg SD 22 healthy male Carbonated With carbonated Cmax reduced 57%, Tmax SMITH et a1.
subjects beverage beverage increased from 0.71 to 1.11 (1993)
h. Bioavailability reduced
28%
Fluvastatin Capsule 20mg SD Healthy male Low-fat evening 4 h after meal 12 h plasma Cmax reduced 44%, Tmax SMITH et al.
subjects meal (6 p.m.) increased 57 %. (1993)
Bioavailability reduced 14%
Fusidate sodium Film-coated 500mg SD Standardized After meal 26 h serum Cmax reduced 26%, Tmax MACGowAN
tablet breakfast increased from 2.2 to 3.2 h. et al. (1989)
AUC marginally
reduced (16%)
H ydroxychloro- Tablet 155mg SD 9 rheumatoid Light breakfast After meal Absorption delayed due to McLACHLAN
quine arthritis patients food to similar extent to and CUTLER
healthy subjects (1991)
Isosorbide-5- Controlled 60mg SD 18 subjects High-fat breakfast Cmax reduced 5%, Tmax KOSOGLOU
mononitrate release tablet increased from 3.1 to 6.5 et al. (1993)
h. AVe increased 9%
Lomefloxacin Capsule 400mg SD 12 healthy High-fat or high'- 200ml water 5 min after meal 48h plasma, Tmax delayed approximately HOOPER et a1.
subjects, 8M, 4F carbohydrate 72 h urine I h, em" and AUC (1990)
breakfast unchanged
Loracarbef Capsule 400mg SD 12 healthy male Standard breakfaste 200ml water 5 min after meal 24 h serum Cmax reduced 29%, Tmax ROLLER et a1.
subjects fasting, 100 and urine increased from 68 to 141 min, (1992)
ml water fed AUC unchanged
Loracarbef Capsule 200mg SD 12 subjects 12 h plasma Cmax reduced and Tmax DESANTE and
delayed. AUC unchanged ZECKEL (1992)
Methotrexate Tablet 7.Smg SD 12 healthy male Standard high-fat 10 min after meal 24h plasma Cmax reduced by 16%, KOZLOSKI et al.
subjects breakfast e Tmax delayed from 1.0 to (1992b)
1.6 h. AUC increased 4.6%
Methotrexate Tablet 1Smg SD 10 patients, SM, Standard French After meal 24h plasma Cmax reduced 31 %, Tmu OGUEyet a!.
SF breakfast' increased from 1.3 to 2.0 h. (1992)
Bioavailability unaffected
Monofluoro- Chewable tablet 10mg SD 8 healthy male Standard meale 200ml Immediately after 48 h plasma Lag time delayed from 4 WARNEKE and
phosphate fluoride subjects fluoride-free meal and urine to 11 min, Cmax reduced by SETNIKAR
with 300mg water 67%. Tmax increased from (1993)
calcium 24 min to 2.4 h. AUe and
urinary excretion not
affected
Moricizine Tablet 2S0mg SD 24 healthy male Standard breakfaste 180ml 30 min after meal 24h plasma Cmax reduced 24%, Tmax PIENIASZEK
subjects orange juice delayed from 0.9 to 1.2 h. et aJ. (1991)
AUC not affected
Nicorandil Tablet 10mg SD 10 healthy male Standard breakfast' - 10 min after meal - Cm~ reduced 13%, Tm~ FRYDMAN
subject increased from 0.38 to (1992)
0.73 h, AUC increased 24%.
Absorption rate consistently
decreased by 90%
Nifedipine 10mg SD 10 healthy subjects High-fat meal 100ml water After meal 24h plasma Cmax reduced by 47%, Tmu RAPIN (1992)
increased from 0.9 to 1.44 h,
AUC unaffected. Under
same study conditions of
nicardipine and nitrendipine
unaffected
Ofloxacin Tablet 200mg SD 7 healthy male 300ml milk 300 ml water Immediately 24 h plasma Cmax reduced 18%, Tmax NEUVONEN and
subjects 300 ml yoghurt' before milk and urine delayed 1 h by yoghurt. KlVlSrti (1992)
or yoghurt AU C unaffected. Milk had
no effect on any parameters
Ofioxacin Capsule 300mg SD 21 healthy male Standard breakfast' - 30 min after food 24 h plasma Cmax reduced 17%, Tmax DUDLEY et a1.
subjects or milk or milk delayed 1 h by food, AUC (1991)
unaffected. Milk had no
effect on any parameters
Table 4. Continued
Drug Dosage form Dose Dosage regimen Subjects Food details Fluid Time interval Sampling Result of food Reference
volume duration administration
Paracetamol Tablet and 500mg SD 12 healthy male 200mlliquid 200ml With liquid diet 12h plasma The two liquid diets did WALTER-SACK
solution subjects formula enriched not differentially affect et al. (1989)
with or depleted of absorption from solution.
dietary fiber' Fiber-depleted diet delayed
absorption from tablets
relative to fiber -enriched diet
Paracetamol Tablet l000mg SD 12 healthy Tswana Cereal, maize, or 200ml water 30 min after meal 12 h serum Absorption delayed to WESSELS et a1.
female volunteers bacon and egg increasing extent by maize, (1992)
breakfastC bacon and egg. and cereal
breakfasts
Penciclovir 250 or SD 12 healthy male 24h plasma Cmax reduced 47% and FOWLES et a1.
(famcic1ovir 500mg subjects 45%, Tm" delayed by 1.7 (1990,1991)
prodrug) hand 1.8 h from 250- and
500-mg doses, respectively,
AUe unaffected
Penciclovir 500mg SD 18 healthy male 2 h before or 24h plasma emax reduced 18%, Tmax FOWLES et aI.
(famcic10vir subjects after meal and urine delayed by 1.1 h (1990, 1991)
prodrug)
Rifabutin Capsule 150mg SD 12 healthy male Standard high-fat 15 min after meal 168hpiasma, Cmaxreduced 17%, Tmax NARANG et a1.
subjects breakfast' 48 h urine delayed 2.4 h, Aue (1992)
unaffected. Urinary
excretion reduced 20%
Salsalate Tablet 1500mg SD 17 healthy male Standard high-fat 6 ounces Immediately after 48h plasma Salsalate mean Cmax reduced HARRISON et al.
subjects breakfastC water meal 17%, Tmu delayed 1 h, AUe (1992)
unaffected. Salicylate
parameters unaffected
Terazocin 2mg SD 12 subjects Standard breakfast' 100 ml water - 30h plasma Mean Cmax reduced 23 %. McNEAL et al.
Tmu delayed 0.9 h, AUe (1991)
unaffected
Terfenadine Tablet 120mg SD 24 healthy male Standard breakfast' 200ml water Immediately after 48 h plasma Mean metabolite 1 Cmax ELLER et at.
subjects meal increased 13%, TmaJ[delayed (1992)
by 0.9 h. AUe unaffected
Theophylline Theo-Dur tablets 400mg SD 10 healthy male High-fat dinner' 6 ounces Immediately after 36h serum Mean Cmax increased 33 %, KANN et al.
subjects water meal, 8 p.m. T ron unchanged, AU e (1989)
increased 6%. Absorption
rate delayed during initial
8 h postdosing
Theophylline Multiparticulate 300mg SD 12 healthy male Standard breakfastC 150ml water Immediately after 36h serum Delayed drug absorption YUEN et a1.
CR pellet subjects meal associated with delayed (1993)
stomach emptying
Tiagabine Tablet 8mg SD 18 healthy male Cmwt reduced 44%, Tmu MENGEL et a1.
subjects increased from 0.9 to 2.6 h. (1991)
AUC unaffected
Topiramate Tablet 100mg, SD 18 healthy male Fatty breakfast 168 h plasma Cm.. reduced 11 % (100 mg) DOOSE et al.
400mg subjects and 13% (400mg), Tmu (1992a)
increased by 1.8 h (100 mg)
and 2.1 h (400 mg), AUC
(0-=) unaffected
Trazodone Capsule 100mg SD 8 healthy subjects, Standard breakfast After meal 26h serum Cmax reduced 22%, T JDBJ. NILSEN and
4M,4F and urine increased from 1.3 to 2.0 h. DALE (1992)
Amount of drug absorbed
unchanged
Valproic acid Tablet 800mg SD 16 healthy male Light meal, heavy" 180 ml water After meal 45 or 48h emu reduced 14% and AVe OHDO et al.
subjects meal plasma 9% by heavy evening meal, (1992)
but not affected by light
evening meal relative to
light breakfast
Vigabatrin Tablet 1000mg SD 24 healthy male 36 h plasma, Cmax reduced 33%, Tmax HOKE et al.
subjects 48 h urine increased from 1.0 to 2.14 h. (1991)
Extent of absorption
unaffected
Zalospirone 20mg SD 24 healthy male High-fat, low-fat After meal 24h plasma Cmu reduced 13% by low-fat KORTH-
subjects breakfast and 31 % by high-fat meals. BRADLEY et a1.
Tmax increased 0,5 hand (1992)
1.2 h. AU C unaffected
Zidovudine 100mg SD 18 asymptomatic High-fat breakfast 30minor3h 10h plasma Cmax reduced 57% (30 min) SHELTON et a1.
HIV -infected after meal and 47% (3 h). Tm .. (1993)
subjects increased by 1 h (30 min)
and 0.6 h (3 h). AUC
reduced 3% (30 min) and
15% (3 h)
Zidovudine Capsule 200mg SD 11 symptomatic Liquid protein meal 150 ml water Immediately after 8h serum emu reduced 32%, TrnaJ. SARAI et a1.
HIV-infected in 220 ml orange meal and urine increased from 49 to 74 min. (1992)
men juicec AVe and renal clearance
unaffected
40
aJ
~ 36 aJ '"0 aJ
S '"0
"C
"C
C '"0
"C
~ 32
.
0
.
OIl OIl
C 0
,;:;. c c
'2 '2
aJ
:2 28 0 .:t: aJ
\J :; ...: >
'"
L.L.I
.~ 24
>..
~ 20
'"'c0"
·s 16
.;.,'" 12
'"
E 8
''""
"6:
4
0
-1 3 7 11 15 19 23 27 31 35 39 43 47
07.00 14.00 21.00 07.00
3T---------------------------------------------------,
~
5
c
0
..
·0
IU
;;
l:
c..
·0
IU
E
'"
IU
E:
0
-1+---~--~--~--_r--_r--~------_,--_,--_,r__,r_--~
o 2 4 6 8 10 12
Time (hr)
delayed but not reduced following rice and bread meals, with rice having a
greater effect than bread (OGUMA et al. 1991). For both meal types, the effect
was greater for a large meal than for a small meal. Similar delays in cefaclor
absorption were observed when drug was taken immediately after a standard
breakfast, resulting in a 51 % reduction in mean peak plasma levels
(BARBHAIYA et al. 1990). Under the same study conditions peak levels of
cefprozil were reduced by only 14% and plasma profiles were similar after
fasting and nonfasting treatments. The different effects on plasma profiles of
cefaclor and cefprozil are shown in Fig. 8. It is claimed that because cefaclor is
absorbed more rapidly than cefprozil under fasting conditions, slight perturba-
tions in stomach emptying and GI motility are more likely to affect the
absorption rate of cefaclor than of cefprozil. Other studies have shown
cefadroxyl to be unaffected by food, similarly to cefprozil, while absorption of
cephalexin is delayed similarly to cefac10r (LODE et al. 1979). In further studies
designed to examine the influence of gastric emptying on cefprozil absorp-
tion, food delayed the cefprozil tmax in plasma from 1.5 to 2h but did not
affect peak cefprozil plasma levels or absorption efficiency. Pretreatment
with metoclopramide reduced the tmax and mean residence time while
propantheline increased the tmax and significantly increased mean residence
72 P.G. WELLING
8 8
7 7
-
E
6
-
E
6
~5 "Ob
.
5
2-
·N
e
0
4 1:i 4
~
~ <II
'!IS"' '"'
!IS
E 3 E 3
'"
!IS '"
!IS
ii: ii:
2 2
o~----~------~----~----~ O~----~------~----~-------i
o 2 4 6 8 o 2 4 6 8
Time (hr)
Fig. 8. Mean plasma versus time concentrations of cefprozil and cefaclor in 12 subjects
following single 250-mg oral doses of cefprozil or cefaclor under fed (e) and fasting
(0) conditions. (From BARBHAIYA et al. 1990)
Time (hr)
Fig. 9. Mean plasma versus time concentrations (±SD) of fluoride in eight subjects
following a single lO-mg oral dose of fluoride as sodium monofluorophosphate fasting
(0) or after a standard meal (e). (From WARNEKE and SETNIKAR 1993)
Food has been shown to decrease the absorption rate of the salicylic acid
derivative salsalate (salicylsalicylic acid) (HARRISON et al. 1992). However, the
effect is modest, and there was essentially no effect on circulating levels of the
major metabolite salicylic acid. A similar modest effect has been reported for
terazosin, whose absorption was moderately delayed by food, giving rise to a
23% reduction in Cm• x and an 0.9-h increase in Tmax (McNEIL et al. 1991).
Consistent with this modest effect, the maximal fall in standing blood pressure
after food was similar to that in the fasting state. Similar minor effects were
observed with terfenadine, with peak levels of the active metabolite decreas-
ing only 13%, with an 0.9-h delay following a standard high-fat breakfast
compared to the fasted state in healthy volunteers (ELLER et al. 1992).
Two studies reporting delayed absorption of theophylline due to food
have contributed further to the wide spectrum of food effects that have been
reported for this compound and its many formulations. Absorption of theo-
phylline from Theo-Dur tablets was delayed to a small extent, but mean peak
levels increased from 4.7 to 6.3mg/ml from a 400-mg postprandial evening
dose compared to the fasting state. While these results are consistent with
those reported previously regarding delayed absorption, the marked increase
in Cmax ' together with observed intersubject variability in serum profiles ob-
tained following evening doses, may have implications for asthmatic patients
with nocturnal asthma who require consistent medication at night. In a second
study, the onset and rate of gastric emptying were both delayed by a standard
meal and the rate of theophylline absorption from a multiparticular controlled
release formulation was also delayed, but to a lesser extent (YUEN et al. 1993).
Calculations of the relative amounts of drug absorbed from regions of the GI
tract showed that, in fasted individuals, 9% was absorbed from the stomach
and 54 % from the small intestine. In fed individuals 14 % was absorbed
from the stomach and 47% from the intestine. Following both treatments
37%-39% of the absorbed dose was absorbed from the colon, showing this to
be an important absorption site, at least for sustained-release products of
theophylline.
Delayed absorption due to food has been reported for the gamma-
aminobutyric acid uptake inhibitor tiagabine (MENGEL et al. 1991). In a study
in healthy male volunteers, peak plasma levels were reduced by 44 % and Tmax
was increased almost threefold under nonfasting conditions compared to fast-
ing conditions. AUC(0-48) values were similar for the two treatments. Similar
observations were made for the structurally novel anticonvulsant topiramate,
although peak plasma levels were reduced only 11 % and 13 % from single 100-
mg or 400-mg doses and Tm • x was increased by only 1 h (DOOSE et al. 1992a).
Absorption efficacy was unaffected by food. Similar modest delays in absorp-
tion have been reported for the serotonin uptake inhibitor trazodone (NILSEN
and DALE 1992). A more complex study on valproate pharmacokinetics, on the
other hand, shed some light on the influence of food on apparent "circadian
rhythm" in absorption of this and possibly other compounds (ORDO et al.
1992). In a study conducted in healthy male subjects, valproate was adminis-
Drug-Food Interactions Affecting Drug Absorption 77
tered following conventional light breakfast and heavy evening meals and also
following identical morning and evening meals. There was no fasting treat-
ment in this study. Following the light breakfast-heavy evening meal doses,
absorption of valproate was delayed following the evening dose relative to the
morning dose, Cmax being reduced by 14% and AUe by 9%. When valproate
was administered following identical morning and evening meals, there were
no differences in the resulting plasma valproate profiles. The different results
in the two treatments were attributed to differences in meal sizes and
the results of the study led to speculation as to what extent different food
effects may have contributed to many "circadian rhythm" effects previously
reported for other drugs. Plasma levels obtained in this study are shown in
Fig. 10.
Different meals also influenced the extent of drug-food interaction with
the serotonin agonist/antagonist zalospirone. In a study in 24 healthy men,
administration of a single 20-mg zalospirone dose after a meal containing 43%
fat resulted in a 31 % reduction in peak circulating drug levels compared to the
fasting state. Administration after a meal containing 19% fat, on the other
hand, resulted in only a 13 % reduction. Peak drug levels were delayed twice as
long after the high-fat meal as after the low-fat meal. AUe values and
incidences of adverse events were similar for all dosage treatments.
Whereas previously cited reports have claimed delayed and reduced
absorption of the AIDS drug zidovudine (see Table 3), other studies have
reported only delayed absorption, with no influence by food on overall
absorption efficacy (SAHAI et al. 1992; SHELTON et al. 1993). In a study in 18
asymptomatic HIV-infected subjects, Cmax was reduced 57% and 47% when
zidovudine was administered 30min and 3h after a high-fat breakfast, with
moderate increases in Tmax , while overall absorption efficacy was unaffected
(SHELTON et al. 1993). In a second study (SAHAI et al. 1992), serum zidovudine
Cmax was reduced 32% and Tmax increased from 49 to 74min when drug
was administered after a liquid protein meal relative to the fasted state in
11 symptomatic HIV-infected men. Again, absolute absorption values
were unaffected. The different results obtained in these studies compared
to those described in Table 3 present another example of the variable
nature of drug-food interactions and the hazards of basing any conclusions
on only one study from one laboratory, using a single set of study
conditions.
100 100
E 90 90
~ A B
2- 80 80
"t:I
'0 70 70
..s
1.1
'0 60 60
.
Q.
-;;; 50 50
..
..s 40
E 40
'"..s 30 30
!
E:
20 20
i !!
10 " 10
0 I I
II
I I
"I
o I I I I I I 1/ T
0 2 3 4 12 24 33 36 o 2 3 4 6 12 24
Time (hr) Time (hr)
Fig.l0A,B. Mean plasma versus time concentrations (±SD) of valproic acid in 16 subjects following single 800-mg oral doses
of valproic acid following a light breakfast (e) or heavy dinner (_) (A) or following a light breakfast (e) or light dinner (_)
(B). (From ORDO et al. 1992)
""d
o
~
tn
t'"
t:
z
Cl
Table 5. Drug-food interactions resulting in increased drug absorption
Drug Dosage form Dose Dosage Subjects Food details Fluid volume Time interval Sampling Result of food administration Reference
regimen duration
Alprazolam Sustained 3mg 29 healthy Standard high-fat _b 1 h before, 48h blood Cmax increased 10%-17% WRIGHT (1992)
SD'
release tablet subjects meal immediately after food. AUC marginally
following, 1 h increased (::;;6%). Psychomotor
after, and 2 h performance decrement
after meal consistent with changes
in blood levies
Amiodarone Solution 300mg SD 10 healthy male Nutrient RealmentylC 600 ml water Drug 24h plasma, Amiodarone absorption PFEIFFER et al.
subjects solution at two administered in 2 h jejunal correlated with lipid (1990)
dilutions infused 120 nutrient solution aspirations absorption. Plasma levels
min at ligament of variable
Treitz
Amocarzine Film-coated 3 mg/kg MDd 20 male Copious breakfast 100 ml water 1 h after meal 10h plasma AUe increased 20%, TrnlU LECAILLON
breakable tablet Guatemalan and urine delayed 2 h, Cmu unaffected. et al. (1991)
patients infected T m" of metabolite CGP 13231
with Onchocerca delayed 2 h
volvulus
Amocarzine Film-coated 1200 mg SD 11 male patients Standard breakfastC 100ml water After meal 48 h plasma, Cmax and AUe of parent LECAILLON
tablet infected with 480rl44h drug increased threefold. et al. (1990)
Onchocerca urine Urinary excretion increased
volvulus from 0.11 % to 0.13% of dose.
C max , AVe and urinary
excretion of metabolite eGP
13231 increased threefold
Astemizole and Capsule 10 mg astemizole, SD 28 healthy Standard meal With meal 96h plasma Cmax for astemizole JALLAD et a1.
Pseudoephedrine 140mg subjects and metabolite (1991)
pseudoephedrine desmethylastemizole
delayed while AUC increased.
Pseudoephedrine unaffected
Atovaquone Tablet 500mg SD 18 healthy male High-fat breakfast' 45 min after 528h plasma C max increased fivefold, AVe ROLAN et al.
subjects meal increased threefold (1994)
Atovaquone Tablet 500mg SD 12 healthy male Toast, toast with 45 min after 336 h plasma C max and AUe both increased ROLAN et a1.
subjects low-fat or high-fat meal with increasing fat meal. (1994)
butter Following fat toast with 56 g
butter. emu and AVC
increased 5.6- and 4-fold,
respectively
Table 5. Continued
Orug Dosage form Dose Dosage Subjects Food details Fluid volume Time interval Sampling Result of food administration Reference
regimen duration
Bay·X·1005 Tablet 500mg SO 4 healthy subjects American breakfast emax and AVe increased BECKERMANN
2·fold and l.4·fold, et al. (1993)
respectively. Tmu reduced
from 4.4 to 2.2 h
Brofaromine Tablet 75mg SO B healthy male Standard breakfast 100ml Immediately 4Bh plasma emax and AVe increased DEGEN et al.
subjects mineral after food 1.2-fold. Tmax reduced from (1993)
water 3 to 2h
Buflomedil Tablet 600mg SD 111In_ 7 healthy male Standard light and 100ml water After meal 25 h serum, Cm8~ and AU C increased WILSON et a1.
labelled subjects heavy breakfasts' scintigraphy approximately 1.2·fold after (1991)
heavy meal relative to light
meal
Cefetamet Tablet 1000mg SO 6 healthy male emu and AVe increased BLOUIN and
pivoxil subjects 1.3·fold. Tmn delayed from STOECKEL
3.0 to 4.Bh (1993)
Cefetamet Tablet 1000 mg SO 16 healthy male Standard breakfast' Various With meal 24h plasma Cmu increased 1.2·fold, AUC TAM et aJ.
privoxi subjects and urine increased 1.1-fold when (1990)
taken 1 h after food compared
to 1 h before
Cefuroxime Tablet 250mg SO B or 12 healthy Cholecystokinin or Hyoscine butylbromide MAcKAyet aJ.
fasting male hyoscine mediated delayed gastric (1991)
subjects butylbromide, no emptying had no effect.
food Cholecystokinin-mediated
increased bile release caused
1.2-fold increases in
C_andAUC
CGP 43371 Capsule BOOmg SO 12 healthy male Standard breakfast C After meal 96h plasma Cmax and AU C increased SUN et al.
subjects 11· and 14·fold, respectively. (1994)
Tmax unaffected
Clarithromycin Suspension 7.5 mglkg SO 24 infants and Milk for patients 125 mg/5 ml 6h plasma Cmax for parent drug and GAN et aJ.
children with <2 years old, milk metabolite increased 1.3- and (1992)
pharyngitis, otitis and hash brown 1.1·fold. AUC increased
media, or skin potatoes for patients 1.4· and 1.1 ·fold
infections ;,2 years old
Clarithromycin Tablet 500mg SD 27 healthy male Standard breakfast 240ml water 30 min after 24h plasma Cmax for parent and metabolite CHU et a!.
subjects start of meal increased 1.5- and 1.1-fold, (1992)
AVC increased 1.25-
and 1.1-fold
Cyclosporine Chocolate 10 mg/kg SD 8 healthy Low-fat and high-fat Midway 24 h blood Based on blood data, GUPTA et al.
emulsion subjects, 4M, 4F breakfast' through high-fat and plasma bioavailability was 23% and (1990)
breakfast 42% after low-fat and high-fat
meals. Based on plasma data
bioavailability was 21 % and
79% after low-fat and high-fat
meals. Distribution volume
and clearance increased with
high-fat meals
Danazol Capsule and 100mg SO 11 healthy Standard breakfast 15 min after 36 h plasma Emulsion increased CHARMAN
lipid emulsion female subjects meal bioavailability fourfold et a!. (1993)
relative to capsule, but
was unaffected by food.
Bioavailability of capsule was
increased threefold by food
Diltiazem Extended 45mg SO 16 healthy male Cmax and AVC increased 37% FRISHMAN
release capsule subjects and 13 %, respectively (1993)
Encainide 35mg MO IS healthy male Standard breakfastC 120ml water Mid-meal 24h serum Bioavailability of encainide HILLEMAN
q8h subjects increased 1.8-fold and that of et a!. (1992)
O-desmethyl encainide 1.3-fold.
3-Methoxy-desmethyl
encainide unchanged, as were
electrocardiograms
Felodipine and Tablet 5mg SO 6 healthy male 250 ml grapefruit 250ml water With water 8 h plasma Felodipine and BAILEyet al.
nifedipine subjects juice or orange juice or juice dehydrofelodipine AVC (1991)
increased 2.5- and 1.7- fold by
10mg SO grapefruit juice. Nifedipine
and dehydronifedipine AVC
increased 1.4- and 1.2-fold.
Orange juice had negligible
effect. Felodipine had greater
effects on diastolic blood
pressure and heart rate when
taken with grapefruit juice
Table 5. Continued
Drug Dosage form Dose Dosage Subjects Food details Fluid volume Time interval Sampling Result of food administration Reference
regimen duration
Felodipine Tablet 5 mg SD 9 healthy male 200 ml grapefruit 200 ml water With water or 8h plasma Cmax increased 2.5- and 2.9- EDGAR et a1.
subjects juice of different juice fold and AUC increased 2.9- (1992)
strengths and 3.3-fold by increasing
strengths of grapefruit juice.
Levels of dehydro-felodipine
similarly increased
Fenretidine Capsule 300mg SD 13 healthy male High-fat breakfast' 200 mI water Immediately 72h plasma Fenretidine Cmax and DOOSE et aI.
subjects after meal AU C increased threefold, (1992b)
metabolite Cmaxand AVe
increased 2.3- and 2.5-fold
Fenretidine Capsule 300mg SD 15 healthy male High-fat, 200ml water lOmin after 72h plasma High-fat meal increased DOOSE et a1.
subjects -carbohydrate or meal bioavailability threefold (1992b)
-protein mealsc relative to carbohydrate meal.
High-protein meal
gave intermediate values
Gepirone Capsule 20mg SD 20 healthy male Standard breakfast 200 ml water 15 min after 48h plasma AVe increased lA-fold. Cmax TAyet al.
subjects meal slightly reduced and delayed (1993)
Itraconazole Capsule 100mg SD 6 healthy male Standard breakfast C 200ml water Immediately 96h plasma Cmax increased 3.4- VAN PEER
subjects after meal fold, AUC 2.6-fold et al. (1989)
Itraconazole Capsule 200mg SD 28 healthy male Standard breakfast C 200ml water Immediately 72h plasma Cm~ increased 2- fold, AUC BARONE et a1.
subjects after meal 1.6-fold. Metabolite Cm.. (1993)
increased l.4-fold, AUC
l.5-fold
Itraconazole and Capsules 100mg 12 healthy Standard light or full 100 ml water 5 min after 96 h plasma Itraconazole bioavailability ZIMMERMANN
fluconazole subjects, 7M, 5F breakfast' meal increased l.4-fold by light et aI. (1994)
for each drug meal, and 1.7-fold by heavy
meal. Fluconazole unaffected
Levodopa Immediate or 100mg IR SD 5 healthy male Light breakfast After meal 8h plasma Absolute bioavailability WILDING et a1.
controlled 200mg CR subjects relative to intravenous dose: (1991b)
release tablet IR fed> IR fasted> CR fed
> CR fasted. Food increased
absorption from both
formulations
Levodopa Variable MD 16 patients with Diet rich in soluble Few min after 6h plasma Total plasma levodopa levels ASTARLOA
idiopathic fiber (DRIF)' fiber supplement increased l.3-fold, levels et al. (1992)
Parkinson's during first h postdose
disease and increased 1.5-fold, after
severe 2 weeks on D RIF
constipation,
6M,13F
5-Methoxy- Tablet 20mg SD 9 healthy Standard breakfast' 100ml water 20-70 min after 10 h plasma C~, ranged from 0 to 88 ng/ml EHRSSON et a1.
psoraien volunteers, meal fasting and 37~141 ng/ml (1994)
IM,8F nonfasting. AVC ranged from
oto 422 ng·h/ml nonfasting
Moclobemide 100mg SD 8 healthy male Standard high- 9 h plasma Trend toward higher absolute A.F.D. COLE
subjects protein breakfastc bioavailability after protein et a1. (1992)
meal (67%) compared to
fasting (58%)
Nifedipine Extended 40mg 20 healthy male Low-fat and high-fat 45 min after 48h blood C max increased 1.8-fold and KLEINBLOESEM
release tablet subjects meal meal 2.4-fold, AVC increased 1.2- et a1. (1993)
fold and 1.2-fold, by low-fat
and high-fat meals,
respectively
Oxcarbazepine Tablet 600mg SD 6 healthy male Standardized high- 100 ml water Immediately 72 h plasma Cm~ and AVC of parent drug DEGEN et al.
subjects fat, high-protein after meal increased 1.7- and 1.2-fold, (1994)
breakfast respectively. Values for
monohydroxy and dihydroxy
metabolites also increased
Oxybutinin Solution 15 mglkg SD 18 healthy male High-fat breakfast With meal Bioavailability YONG et al.
subjects increased 1.3-fold (1991)
Phenytoin Powder 5 mglkg 4 healthy male Low-fat and high-fat 200ml water Immediately 34h plasma Cmax increased 1.5- and 2.2- HAMAGUCHI
subjects breakfast' after meal fold by low-fat and high-fat et a1. (1993)
meals, respectively. AVe
increased 1.5- and 1.9-fold
Progesterone Capsule 200mg MD 15 healthy Standard breakfast' Immediately 24 h plasma On day 1, Cmaxincreased SIMON et a!.
5 days postmenopausal after or 2 h on day 1, 10 5-fold and AVe 2-fold. (1993)
women before h plasma on Similar effects on day 5
day 5
Repirinast Tablet 300mg SD 12 healthy male High-carbohydrate l00ml water Immediately 72 h plasma Cmu and A V e increased SCHAEFER
subjects or high-fat breakfast' after meal 1.6- and 1.9-fold by high- et a1. (1993)
carbohydrate breakfast and
3.2- and 2.4-fold
by high-fat breakfast
Sparfloxacin 300mg SD 3 healthy subjects ~ Cm~ increased 1.5-fold, AVC TANIMURA
1.2·fold, and urinary excretion et a1. (1991)
1.4-fold
S-1108 200mg SD 6 healthy male Japanese breakfast l00ml water Immediately lOh blood, Cm~ increased l.l-fold, AVe SAITO (1993)
subjcets -500 Kcal after meal 24 h urine 1.4-fold
S-1108 l00mg SD Healthy male C max increased l.4·fold, AVe NAKASHIMA
subjects 1.5·fold. T max increased from et a1. (1993)
1.4 to 2.5 h
Table 5. Continued
Drug Dosage form Dose Dosage Subjects Food details Fluid volume Time interval Sampling Result of food administration Reference
regimen duration
Theophylline Capsule 900mg SD 20 healthy High-fat breakfast' 15 min after 72 h serum Cmax increased 1.6-fold, COOK et al.
subjects meal AUC 1.2-fold (1990)
Ticlopidine Tablet 250mg SD 12 healthy male High-fat breakfast' 200ml water 30 min after 24h plasma Cm~andAUC SHAH et al.
subjects meal increased 1.2-fold (1990)
Tramadol Tablet 100mg SD 18 healthy male High-fat breakfast Immediately 36 h plasma C_ and AUC of tramadol LIAO et a1.
subjects after meal increased 1.2- and l.l-fold, (1992)
respectively. Metabolite levels
only moderately affected
Vanoxerine Tablet 100mg SD 12 healthy male High-fat or low-fat 100 ml water After meal C_ increased 1.5-fold by INGWERSEN
subjects breakfastC high-fat meal, unaffected et al. (1993)
by low-fat meal. AUC
increased 1.8-fold and 3.6-fold
by low-fat and high-fat
meals, respectively
Vinpocetine Tablet 40mg SD 8 healthy Standard breakfast, 150ml 10 min before 12 h serum Cmax increased 1.6-, 2.3-, and LOHMANN
subjects, 4M, 1350 Kcal mineral start of meal, 1.9-fold while AUC increased et al. (1992)
4F water 10 min after 1.6-,1.7-, and 2.0-fold when
start of meal, drug was taken 10 min before,
20 min after 10 min after starting, and 30
termination of min after finishing meal
meal
Zalospirone 10mg MD 2418- to 45- Medium-fat meal 30 min after Cmax and Ave increased KLAMERUS
q8h year-old and 24 meal twofold. Elderly subjects gave et al. (1993)
" 65-year- higher levels than younger
old male and subjects
female subjects
566C80 Tablet 500mg SD 18 healthy male 45 min after Cmax increased 5A-fold, AVe ROLAN et a1.
subjects meal 3.3-fold (1992)
566C80 Tablet 500mg SD 12 healthy male Toast, toast plus 28 emax increased 2.5- and 3.2- ROLAN et al.
subjects g butter (LOFAT), fold after LOFAT and (1992)
toast plus 56 g butter HIFAT, respectively,
(HIFAT) compared to plain toast.
again, not only the broad spectrum of effects that may result from drug-food
interactions, but also their frequent unpredictability. The compounds in this
section tend to be lipophilic and poorly water soluble, but this is not always the
case. Similarly some interactions are relatively trivial and do not warrant
further discussion, while others are substantial and likely to be clinically
significant.
Amiodarone is an amphiphilic substance and its bioavailability is ex-
tremely variable (LATINI et al. 1984; RIVA et al. 1982). In order to examine its
absorption in the presence of food, amiodarone was administered into the
jejunum together with two nutrient solutions, one 3.3 Kcal/min and the other
1.3 Kcal/min, with the former containing total lipid and caloric load 2.5 times
greater than the latter (PFEIFFER et al. 1990). Based on intestinal testing,
absorption of amiodarone correlated significantly with lipid absorption rate.
However, plasma Cmax and AVC values were extremely variable and tended to
be higher from the 1.3-Kcal/min infusion. These seemingly paradoxical results
were attributed to wide fluctuations in amiodarone pharmacokinetics, distri-
bution, and metabolism.
Conflicting results from drug-food interactions have been reported with
the onchocerciasis agent amocarzine (CGP 6140) (LECAILLON et al. 1991). In
20 male Guatemalan patients who received a 3-mg/kg oral dose, systemic
availability was increased 20% when drug was taken with a "copious" break-
fast compared to fasting. In a similar study, when the dose was increased to
1200mg, both the peak plasma levels and systemic availability of amocarzine
were increased approximately threefold when drug was given after a meat and
noodle standard breakfast, relative to fasting (LECAILLON et al. 1990). The
AVC of the metabolite CGP 13231 also increased threefold in this study,
although it was unaffected at the lower dose. Mean plasma levels of
amocarzine and its metabolite CGP 13231 are shown in Fig. 11.
The considerable increase in absorption due to food after the high dose of
amocarzine may be related to a greater degree of solubilization after the meal
or to decreased presystemic metabolism. The nature of the food in the lower-
dose study was not described, but is likely to have been similar to that used in
the high-dose study. A controlled high-dose, low-dose study would provide
useful mechanistic information on this interaction. Substantially increased
absorption due to food has been reported for the lipophilic antiprotozoal
agent atovaquone (ROLAN et al. 1994). In healthy male volunteers, peak
atovaquone plasma· levels increased over fivefold while systemic
bioavailability increased over threefold when drug was given 45 min after a
Western-style high-fat breakfast compared to fasting. Mean plasma profiles
obtained over 48 and 528h are shown in Fig. 12.
Complementary studies using meals with different fat content, aqueous
suspensions, and oily emulsion vehicles, and also pre dosing with a cholecysto-
kinin octapeptide, led to the conclusion that the observed food effect with
atovaquone was probably due to the combined effects of bile release and also
increased solubility due to a direct effect of the fatty meal.
86 P.G. WELLING
15000
12000 A
9000
6000
S0 3000
E
E
c 6 12 18 24
.
0
~
C
~
15000
<oJ
C
0 12000 B
u
9000
6000
3000
I
6 12 18 24
Time (hr)
Fig.llA,B. Mean plasma versus time concentrations (±SD) of CGP 6140 (A) and its
N-oxide metabolite CGP 13231 (B) in 11 patients following a single 1200-mg oral dose
of CGP 6140 fasting (0) or following a large breakfast (e). (From LECAILLON et al.
1990)
4
E
~
~
cu 3 0
c::
0 84 168 252 336 432 528
j
cr
-
t':S
:>
0
t':S 2
t':S
E
<I>
t':S
'2i:
1
o~--------~--------~~--------~--------~
o 12 24 36 48
Time (hr)
Fig. 12. Mean plasma versus time concentrations of atovaquone in 18 subjects follow-
ing a single SOO-mg oral dose of atovaquone fasted (0) or 4Smin after a high-fat meal
(e). (From ROLAN et al. 1994)
tained with cefetamet pivoxil administered under fasting and nonfasting con-
ditions, and with different fluid volumes, are shown in Fig. 13. The lack of
effect of fluid volume in this study may have been due to the relatively large
fluid volumes used, 250ml and 450ml. However, the lack of a food effect when
drug was administered directly with the meal was unexpected. A proposed
explanation is that while drug was taken with water 1 h before and 1 h after
breakfast, it was taken with the tea or coffee provided for the group that
received the drug with food. Previous studies have shown that bioavailability
of cefuroxime is increased when taken with food (WILLIAMS and HARDING
1984). In an attempt to identify the mechanism of this interaction, cefuroxime
was administered to healthy male subjects 30 min before intravenous hyoscine
butylbromide or immediately before intravenous cholecystokinin 8 (MAcKAY
et al. 1991). Hyoscine butylbromide had no effect on cefuroxime absorption
while cholecystokinin resulted in a 20% increase in cefuroxime emax and AVe
values. These results led to the conclusion that bile release, but not gastric
emptying, may be at least partially responsible for increased cefuroxime ab-
sorption in the presence of food.
Possibly the most remarkable food effect reported during the review
period involved the lipophilic hypolipidemic compound eGP 43371 (SUN et al.
1994). Administration of single 800-mg capsule doses of eGP 43371 after a
88 P.G. WELLING
7
6 Cl"',Ci
~
E 5 .'0' ......
II
-
II
B
~ I
\\
\\
....
\\
4
-
Q,I \\
E
~
\\
\\
JQ,I! 3
\J
51...
'\
~
E 2
<II
\~
~
ii: .... ....
1
0
0 2 4 6 8 10 12
Time (hr)
Fig. 13. Mean plasma versus time concentrations of cefetamet in 12 subjects following
a single lOoo-mg oral dose of cefetamet pivoxil under fasted (-) or fed (---) conditions
with 250ml (0) or 450ml (0) water. (From TAM et al. 1990)
2.5
1 2.0
~
,...
I'-. 1.5
..,
M
M
=- 1.0
1..7
u
~
E
<II
~
ii: 0.5
0.0
0 12 24 36 48 60 72 84 96
Time (hr)
Fig.14. Mean plasma versus time concentrations of CGP 43371 in 12 subjects following
a single Boo-mg oral dose of CGP 43371 as a dispersion (6) or capsule (0) under fasting
conditions or as a capsule after a standard meal (e). (From SUN et al. 1994)
standard breakfast caused an ll-fold increase in peak plasma drug levels and
a 13-fold increase in overall bioavailability relative to the fasting state. Plasma
levels from this study are shown in Fig. 14.
It is proposed that, as CGP 43371 is absorbed mainly from the ileum,
delayed gastric emptying would enable more compound to disintegrate and
Drug-Food Interactions Affecting Drug Absorption 89
dissolve before reaching this absorption site. It is further proposed that CGP
43371 dosage should be modified depending on dosing relative to food intake.
This study provides a truly dramatic example of the extent to which drug-food
interactions can influence circulating drug profiles, with obvious clinical
implications.
Similar food effects were observed in infants and adults for the new
macrolide antibiotic clarithromycin (CHU et al. 1992; GAN et al. 1992). Follow-
ing a 7.5-mg/kg dose to infants and children aged 6 months to 10 years with
various infections, either fasting or after milk and/or hash brown potatoes,
peak plasma levels were 4.6,ug/ml and 3.6,ug/ml after nonfasting and fasting
doses, respectively (GAN et al. 1992). Although these increases were modest,
systemic availability increased by 40%, indicating better overall absorption
with food. In the study in adults, food taken immediately before a 500-mg
clarithromycin dose increased the extent of absorption by approximately 25%
(CHU et al. 1992). In both of these studies plasma levels of the major active
metabolite 14-hydroxyclarithromycin were moderately increased by approxi-
mately 10%. Different results were obtained from blood and plasma analysis
regarding the effect of low-fat and high-fat meals on cyclosporine absorption
(GUPTA et al. 1990). Based on plasma data, cyclosporine systemic availability
was 23% and 42% after low-fat and high-fat meals, respectively. Based on
blood data, relative values were 21 % and 79%. Thus, the apparent increase in
cyclosporine bioavailability appears to depend on the sample matrix. This in
turn is probably related to differential penetration of cyclosporine into red
cells depending on the amount of absorbed fat. Plasma to blood cyclosporine
clearance ratios were 1.8 and 1.3 after high-fat and low-fat meals, respectively.
Other studies have shown that high-fat meals increased cyclosporine
clearance, but not mean residence time, based on either blood or plasma
measurements (GUPTA and BENET 1990).
Absorption of the heterocyclic steroid derivative danazol (CHARMAN et al.
1993) and also the retinoid fenretinide (DOOSE et al. 1992b) is substantially
increased by food. In the case of danazol, systemic availability from a capsule
dose was increased over threefold by food in healthy female subjects, resulting
in mean peak plasma levels of 37 and 101 ng/ml, after fasting and fed doses,
respectively. In the case of fenretidine, bioavailability and peak plasma levels
both increased threefold following a high-fat meal compared to fasting (DOOSE
et al. 1992). Administration of fenretidine in an oil suspension to fasting
subjects yielded intermediate values. Further examination of the effect of meal
composition showed that a high-fat meal resulted in plasma fenretinide
bioavailability three times greater than a carbohydrate meal, with a high-
protein meal yielding intermediate results. Mean plasma profiles obtained in
these studies are shown in Figs. 15 and 16. These results give rise to the
recommendation that fenretinide be given with food in order to optimize
potential therapeutic benefit.
A specific drug-food interaction has recently been identified between a
component in grapefruit juice and the calcium antagonist felodipine (BAILEY
90 P.G. WELLING
600
500
E
~
-=-
~
400
~
·2
;:; 300
~
i:
~
til 200
E
'"til
E: 100
0
0 12 24 36 48 60 72
Time (hr)
600
500
~
-=-
~
400
·c
~
.
;:;
~
c
300
~
til 200
E
'"til
E: 100
0
0 12 24 36 48 60 72
Time (hr)
et al. 1991; EDGAR et al. 1992). In a study in six men with borderline hyperten-
sion, felodipine and dehydrofelodipine systemic availability increased 2.5- and
1.7-fold, respectively, when felodipine was taken with two 2S0-ml double-
strength grapefruit juices, relative to water (BAILEY et al. 1991). Under the
same conditions, plasma levels of nifedipine and dehydronorfedipine in-
creased 1.4- and 1.2-fold. The results with felodipine were reproduced in
another study in nine healthy middle-aged men (EDGAR et al. 1992). The
interaction with grapefruit juice, which is believed to be a class effect for the
Drug-Food Interactions Affecting Drug Absorption 91
o~--------~----------------~------~
o 10 26 34
Time (hr)
Fig. 17. Mean plasma versus time concentrations (±SE) of phenytoin in four subjects
following a single 5-mglkg oral dose of free acid phenytoin powder with large particle
size fasting (0), and after low-fat (e) and high-fat (.) meals. (From HAMAGUCHI et al.
1993)
Drug-Food Interactions Affecting Drug Absorption 93
related to increased splanchnic blood flow, may have contributed to the food
effect in the other subjects.
Absorption of the nootropic agent vinpocetine and also the 5-HT1• partial
antagonist zalospirone is modestly increased by food (LOHMANN et al. 1992;
KLAMERUS et al. 1993). Administration of vinpocetine tablets 10min before
and 10min after starting and 30min after a standard 1350-Kcal breakfast
increased systemic availability 1.6-, 1.7-, and 2.0-fold relative to fasting. Peak
plasma levels were affected similarly. Mean plasma profiles in eight healthy
male and female subjects in this study are shown in Fig. 18. Peak plasma levels
and areas under plasma curves of zalospirone were increased approximately
lA-fold by food in both young (18-45 years old) and elderly (~65 years old)
subjects (KLAMERUS et al. 1993). In addition, plasma levels were almost
doubled in elderly subjects relative to young subjects. The results of these
studies led to the recommendation that both vinpocetine and zalospirine be
taken with or after meals.
The last drug considered in this category illustrates again the dramatic
positive effect that food can have on circulating drug profiles. In a study in 18
healthy men, the systemic availability of a novel anti protozoal agent 566C80
was increased 3.3-fold, while e m• x was increased SA-fold when administered
30
E
~ 20
5
.c
.;:
ClJ
U
0
c..
c
.;;:
!U
E
<n
!U 10
E:
2 3 4 5 6
TIME (hr)
Fig. 18. Mean plasma versus time concentrations of vinpocetine in eight subjects
following a single lO-mg oral dose of vinpocetine fasting (0) and lOmin before (1:.),
10min after (e), and 30 min after (.) starting a standard breakfast. (From LOHMANN et
al. 1992)
Drug-Food Interactions Affecting Drug Absorption 95
after food (HUGHES et al. 1991). In attempts to elucidate the mechanism of this
interaction, 566C80 was given fasted and with meals of varying fat content, as
an aqueous suspension, and as a oily emulsion. In a related study 566C80 was
given after an infusion of cholecystokinin octapeptide (CCK-OP) (ROLAN et
al. 1992). Pharmacokinetic results from these studies led to the conclusion that
increased absorption of 566C80 after food could be quantitatively accounted
for by dietary fat and that stimulation of bile secretion may be a small compo-
nent of the food effect.
Naproxen Tablet 500mg SD 6 healthy Low-fat meal' - At beginning 48h plasma AUC(O-2h) LAFONTAINE
subjects of meal increased. Fluid et al. (1990)
volume may have
contributed
Valproate sodium Sustained 333mg valproate SD 12 healthy Standard 30ml water Midpoint of 72h plasma Cm~ and 6h ROYER-MoRROTT
and valproic acid release sodium, 145 mg female breakfast' meal absorption et al. (1993)
formulation valproic acid subjects increased 1.2-fold.
Systemic
availability
unchanged
• Single dose.
b Data not available.
, Meal described.
'"
o
~
E
z
Cl
Drug-Food Interactions Affecting Drug Absorption 97
50
40
<:
z 30
~
..b
os
E
'I>
os 20
E:
10
0
0 10 20 30 40 50 60 70 80 90 100
TIME (hr)
40
B
<: 30
z
~
..b
os
E 20
'I>
os
E:
10
0
0 12 24 36 48 60 72
TIME (hr)
Fig. 19A,B. Mean plasma versus time concentrations of the active metabolite of
nabumetone in A 12 subjects following a single lOOO-mg oral dose of nabumetone
fasted (0) or after a meal (e), and in B 15 patients following a single lOOO-mg oral dose
of nabumetone administered with water (0), milk (e), and aluminum hydroxide (.).
(From HYNECK 1992).
Alprazolam Matrix SR Img SO, 21 healthy Standard 170ml water 30min after 36h plasma Cmax increased 12 %. ELLERand DELLA-
tablet subjects, high-fat starting meal AUC and tm~ COLETIA (1990)
12M,9F breakfast b unchanged
Amlodipine Capsule lOmg SO 12 healthy male Standard 150ml water Within 30 min 192h plasma No effect on plasma FAULKNER et a1.
subjects breakfastb after completing levels (1989)
meal
Bambuterol 20mg MO'1 22 healthy Dinner Immediately No effect on plasma ROSENBORG
tablet each subjects after meal levels or urinary et al. (1991)
evening, excretion of
14 days bambuterol or active
metabolite terbutaline
Bisoprolol and Combination 10 mg bisoprolol, SO 16 healthy With meal 48h plasma Tm.. for MURALIDHARAN
hydrochlorothiazide tablet 6.25mg subjects and urine hydrochlorothiazide et al. (1992)
hydrochlorothiazide increased 20%
otherwise no effect on
plasma or urine
parameters
Brofaramine 150mg/day, 14 days MO 10 subjects Cheese or 25-75 mg - Blood Tyramine caused 40 BlECK et aL
tyramine pressure mm increase in blood (1993)
determination pressure. Mean
increase only 11 mm
with cheese
Bromocriptine Tablet 7.5mg SO 7 healthy male Standard breakfastb 100ml water After meal 14h plasma Cmax decreased 10%, KOPITAR et a1.
subjects AUC unchanged (1991)
Carbamazepine Tablet 200mg SO 12 patients with Very low protein AUC decreased 10%, BANNWARTH
mild to advanced diet Tmax reduced from et al. (1992)
chronic renal 8.5 to 6 h, emu
failure unaffected
Cardizem Capsule 360mg SO 18 healthy 36h plasma Diltiazem emax Yu et al. (1992)
subjects reduced 12%,
AU C unchanged.
Deacetyldiltiazem
Cmax increased 11 %,
AU C unchanged
Cefetamet pivoxil Syrup 385mg SO 18 healthy male Standard breakfast' 10ml syrup With meal 15h plasma, Plasma and urine DUCHARME
subjects 24h urine parameters for syrup et al. (1993)
unaffected.
Bioavailability from
syrup lower than
from tablet
Table 7. Continued
Orug Dosage from Dose Dosage Subjects Food details Fluid volume Time interval Sampling Result of food Reference
regimen duration administration
Cimetitine and Tablet 400 mg cimetidine SO 18 healthy male Standard breakfastb 100ml water 15min after 8h plasma No significant effect DESMOND et a1.
ranitidine 150 mg ranitidine subjects starting meal on plasma levels of (1990)
either compound. In
fasted subjects antacid
treatment reduced
plasma levels of
cimetidine and
ranitidine. In fed
subjects antacid had
no effect on either
drug
Cyclosporine Solution 3.51 ± 1.35 MO 14 renal Moderate and 125ml Immediately 12h plasma Cm"andAUC HONCHARIK
mg/kg/day (bj.d.) transplant trace-fat breakfastb orange juice after meal unaffected by trace- et aJ. (1991)
recipients, 9M, fat and moderate-fat
5F meals. Tmax prolonged
from 3.3 to 5.1 h by
moderate-fat meal
Cyclosporine Capsules -6mg/kg SO 11 healthy male Standard light 150ml water With meal 32h plasma Systemic availability LINDHOLM et aI.
subjects breakfast'. also bile unaffected by food (1990)
acid tablet but Cmax reduced
17%. Addition of
bile acid tablets
increased availability
25%
Diazepam, ethinyl 5mg SO 8-10 healthy Olestra or 6 ounces With diet Plasma up to No significant ROBERTS and LETT
estradiol, 0.07mg subjects triglyceride onb water 48h differences in (1989)
norethindrone, 1.0mg absorption when
propranolol 20mg administered with
olestra, triglyceride
oil, or water, except
Tmax for diazepam
prolonged with
triglyceride oil
Diazepam Conventional 60mgCR SO 24 healthy male Standard breakfast 200ml water 30 min after 24h plasma Food had no effect Du SOUICH et a!.
tablet and 120mgSR subjects starting meal on plasma levels of (1990)
slowwrelease diazepam and its
capsule metabolites from
either formulation,
but SR formulation,
increased systemic
availabilty 69%
relative to
conventional tablet
E2020 2mg SD 12 healthy male Standard breakfast Within 30min 168h plasma Rate and extent of MIHARA et a1.
subjects after meal absorption unaffected (1993)
Fluvoxamine Tablet SOmg SD 12 healthy Standard breakfastb 240ml water 15 min after 72h plasma Rate and extent of VAN HARTEN
subjects, 8M, 4F or orange starting meal absorption unaffected et a1. (1991)
juice
Ibuprofen 600mg SD 11 healthy male Standard breakfastb 200mi water 30 min after lOh plasma Plasma levels of S(+) LEVINE et a1.
subjects starting meal ibufrofen higher than (1992)
those of R(-)
ibuprofen under
fasting and nonfasting
conditions. Food
modestly reduced
Cm" of S(+) and
R(-), but systemic
availability, and
isomer ratios
unaffected
Levodopa Solution 125mg SD 8 healthy male 10.5 g or 30.5 g 100ml water 15 min after 8h plasma Meal with 30.5 g ROBERTSON
subjects protein mealsb meal protein had no effect et a1. (1991)
on levodopa
absorption,
Absorption was
reduced 10%, and
C_ by 26% by low-
protein meal
Methotrexate 9.S ± 5.8 mg/week SD 11 patients with Standard meal 24h plasma Rate and extent of PHELAN et a1.
rheumatoid absorption unaffected (1991)
arthritis
Metoprolol Controlled 400mg SD 18 healthy male Standard high-fat Tendency to increase SANDBERG et a1.
succinate release tablet subjects breakfast Cm" and AUC, but (1990)
not significant.
Similar emax values
Metoprolol Controlled 50mg SD 12 healthy Standard Plasma levels SANDBERG et a1.
succinate release tablet subjects carbohydrate unaffected by food (1988)
breakfastb
Morphine sulfate Sustained 30mg 9.12h MD 24 healthy male Standard caffeine- 180ml water Immediately 84hplasma Rate and extent of BASS et al. (1992)
release tablet subjects free meals after meals absorption unaffected
Mosapride citrate Tablet lOmg SD 10 healthy male Sandwiches and 200ml water 30 min after 24h plasma C~. and AUC not SAKASHITA et a1.
subjects 200 ml orange meal significantly affected. (1993)
juice Tmax increased from
0.6 to 0.9h
Moxonidine Tablet O.2mg SD 18 healthy male Standard breakfast 200ml water 30 min after 24h plasma Crnax decreased THEODOR et a1.
subjects meal and urine 14%, Trnax (1992)
marginally increased.
Systemic availability
and urinary excretion
unchanged
Table 7. Continued
Drug Dosage form Dose Dosage Subjects Food details Fluid volume Time interval Sampling Result of food Reference
regimen duration administration
a Single dose.
b Meal described.
C Multiple dose.
104 P.G. WELLING
::::;-
4
~ A
s<II
.is.. 3
c
:cQ
E 2
-<<U
E
c::'"
<U
10
::::;-
~
E
8
B
S
<II
c
.is.. 6
:cQ
E
-<<U 4
E
<II
<U
c:: 2
tered under fed conditions. This is attributed to the lack of effect by food on
the bioavailability of cefetamet pivoxil from the syrup formulation. While the
mechanism for the different responses of cefetamet pivoxil tablets and syrup
to food is unknown, it is claimed that plasma profiles of unbound cefetamet are
adequate to treat susceptible organisms (DUCHARME et al. 1993). Absorption
of both cimetidine and ranitidine was unaffected when they were administered
to healthy male subjects after a standard breakfast compared to fasting
(DESMOND et al. 1990). In the fasted state, absorption of cimetidine was de-
creased 24%, and ranitidine 59%, when these compounds were taken together
with an antacid. In the fed state, however, co administered antacid did not have
the same effect on cimetidine or ranitidine absorption. It is proposed that the
antacid effect seen in the fasting state is related to impaired tablet dissolution
and to drug binding to unabsorbed antacid. Abolition of this effect by food
may be due to competition for drug-binding sites on the antacid. However, this
binding must be weak because of the lack of effect by food on drug absorption
in the absence of antacid.
Data on the effect of food on cyclosporine absorption are conflicting. In a
study cited in Table 5, bioavailability from a chocolate emulsion was 23% and
42% after low-fat and high-fat meals, respectively (GUPTA et al. 1990). Other
studies have reported no significant change (LINDHOLM et al. 1990) or de-
creased cyclosporine absorption with food (KEOWN et al. 1982). Significant
increases in cyclosporine absorption with food have been reported in renal
transplant patients (PTACHCINSKI et al. 1985). Two further studies have re-
ported little effect by food on cyclosporine absorption (HONCHARIK et al.
1991). In the first study, cyclosporine was administered to 14 renal transplant
patients immediately following a moderate- or trace-fat breakfast. Neither
meal had any significant effect on cyclosporine pharmacokinetic parameters.
Mean Cmax values were 410, 346, and 365ng/ml, and mean AVe values were
2115, 2085, and 2145 ng·h/ml following fasting, moderate-fat, and high-fat
treatments at an average cyclosporine dose of 3.51 mg/kg per day. In the
second study, conducted in healthy male subjects, a standard light breakfast
caused a 17% reduction in mean peak plasma cyclosporine levels, but had no
effect on area under plasma profiles, with mean values of 7283 and 7453ng·h/
ml from a 6-mg/kg dose to fasted and fed subjects, respectively (LINDHOLM et
al. 1990). Addition of bile salts to the nonfasted treatment, on the other hand,
caused mean Cmax values to increase 1.1-fold, and overall bioavailability
1.2-fold, relative to fasting. These results suggest that, in this study at least,
bile acid formation is an important determinant of cyclosporine absorption.
This observation does not explain the various outcomes of cyclosporine-food
interactions obtained in other studies.
In an attempt to further elucidate the mechanism of drug-food interac-
tions, and to identify an appropriate surrogate test meal, the absorption of
some widely divergent compounds, diazepam, propranolol, norethindrone,
and ethinyl estradiol, was examined in the presence of water, triglyceride oil,
and a sucrose polyester, Olestra (ROBERTS and LEFF 1989). The study was
106 P.G. WELLING
30
::::i'
E
~
E 20
Q.I
:cQ.
C
0
-:;
to 10
E
'"
to
c;:
72 76 80 84
TIME (hr)
Fig. 21. Mean plasma versus time concentrations of morphine during the 72- to 84-h
dosing interval in 24 subjects following repeated 30-mg q 12h oral doses of morphine
sulfate as sustained release tablets administered under fasting (0) and fed (e) condi-
tions. (From BASS et al. 1992)
108 P.G. WELLING
3.0
2.5
I
~
2.0
c
·u
'><"
0
0;: 1.5
'"
E
<II
I-
'"
E
<I>
1.0
'"
~
0.5
0.0
0 12 24 36 48
Time (hr)
Fig. 22. Mean plasma versus time concentrations of temafloxacin in 18 subjects follow-
ing a single oral dose of 1 x 400-mg tablet fasting (0),2 x 200-mg tablets fasting (0),
and 1 x 400-mg tablet after food (e). (From GRANNEMAN and MUKHERJEE 1992)
Drug-Food Interactions Affecting Drug Absorption 109
pellets in a single tablet gave rise to absorption profiles that were unaffected by
food (WILDING et al. 1992). Healthy volunteers received 600mg drug in the
fasting state and also after standard light and heavy breakfasts. Gamma
scintigraphy measurements showed that stomach emptying of 152Sm-I abeled
pellets was delayed by both light and heavy meals. Despite the delayed stom-
ach emptying, plasma profiles of tiaprofenic acid were almost superimposable
after fasting and fed treatments.
Although food has recently been shown to delay (KANN et al. 1989; YUEN
et al. 1993) and also to increase (COOK et al. 1990) absorption of theophylline
from controlled release formulations, other studies have shown little effect.
The rate and extend of theophylline absorption from Monospan capsules, in
which drug is contained in I-mm-diameter beadlets, were almost identical
when administered to healthy volunteers fasting or immediately after a high-
fat breakfast (HARRISON et al. 1993). Similarly, the systemic availability of
theophylline from Theo-24 capsules was unaffected by enteral liquid feeding
in healthy male subjects (PLEZIA et al. 1990). Earlier reports on the lack of
effect by food on theophylline absorption from Uniphyl tablets during chronic
evening dosing were confirmed in a subsequent study which monitored both
the serum drug levels and also clinical efficacy in patients with chronic airways
obstruction (ARKINSTALL et al. 1991). Following mean daily (evening) theo-
phylline doses of 818mg, peak plasma theophylline concentrations were
14.4.ug/ml with food and 13.1.ug/ml fasting. Mean trough levels were 7.4 and
6.9.ug/ml. There were no treatment-related differences in pulmonary function
tests including spirometry, asthma symptom scores, side effects, or the need
for beta-agonist inhalers.
The last compound to be discussed in this section is of interest largely
because of the divergent results obtained regarding food interactions with
controlled release preparations. Verapamil absorption was previously shown
to be reduced by 30% and peak serum levels by 48% when a sustained release
tablet was taken with food (HOON et al. 1992). However, different results were
obtained when verapamil was administered in a different sustained release
formulation. Twelve healthy male volunteers received single 200-mg ve-
rapamil doses in a newly marketed sustained release formulation employing
rate-controlling technology in the form of a capsule, either fasting or 10min
after a standard breakfast (DEVANE et al. 1990; DEVANE and KELLY 1991).
Plasma profiles from the fed and fasting treatments, together with verapamil
profiles from a conventional 80-mg dose administered every 8h are shown in
Fig. 23. Plasma profiles of verapamil following the sustained release capsule
were superimposable in the fed and fasting states. Plasma profiles of the
metabolite norverapamil were similarly unaffected by food. Mean peak
plasma verapamil plasma levels were 77 ng/ml following both sustained release
treatments, compared to 95 ng/ml following the conventional tablet. Areas
under plasma verapamil concentration versus time curves from zero to infinite
time were 1541 and 1387ng·h/ml from fasted and fed sustained release doses,
respectively, compared to 1370ng·h!ml from the conventional tablet. The
110 P.G. WELLING
80
70
::; 60
~
S 50
·e
tel
c.. 40
~
III
>
tel 30
E
'"
tel
li: 20
10
12 24 36 48
Time (hr)
divergent results obtained from these studies are most likely related to the
formulations used. Release of drug from membrane controlled release formu-
lations, including those using osmotic pump technology, are generally less
sensitive to the gastrointestinal environment, including the presence of food,
than other types of controlled release formulations. The advantage of a rate-
controlling membrane dosage form for verapamil was further demonstrated in
a study in which drug was administered as extended release pellets in the
fasting state and also sprinkled on apple sauce. There were no significant
differences in plasma verapamil or norverapamil profiles between the fasting
and apple sauce treatments (KOZLOSKI et al. 1992a).
I. Conclusions
The number of articles published during the 5 years covered by this review
illustrates the high level of interest, from scientific, regulatory, and clinical
perspectives, on interactions between drugs and ingested food influencing
drug absorption. As in the initial review on this topic (WELLING 1977), drugs
and drug formulations continue to fall naturally into four major categories of
those whose absorption is reduced, delayed, increased, or not affected by food.
In this review, a fifth category of drugs whose absorption is accelerated but not
increased by food was identified.
Grouping of drugs and formulations in this way is entirely empirical. The
interactions could have been grouped differently, for example, among thera-
Drug-Food Interactions Affecting Drug Absorption 111
References
Albertioni F, Juliusson G, Liliemark J (1993) On the bioavailability of 2-chloro-2'-
deoxyadenosine (CdA); the influence of food and omeprazole. Eur J Clin
PharmacoI44:579-582
Arkinstall WW, Brar PS, Stewart JH (1991) Repeated dosing of Uniphyl tablets under
fed and fasting conditions: comparison of serum theophylline levels, pulmonary
function and asthma symptoms. Ann Allergy 67:583-587
112 P.G. WELLING
Chu S-Y, Park Y, Locke C, Wilson DS, Cavanaugh JC (1992) Drug-food interaction
potential of clarithromycin, a new macrolide antimicrobial. J Clin Pharmacol
32:32-36
Clements JA, Heading RC, Nimmo WS, Prescott CF (1978) Kinetics of acetaminophen
absorption and gastric emptying in man. Clin Pharmacol Ther 24:420-431
Cole AFD, Baxter JG, Jackson BJ, Hew-Wing P, Guntert TW, Lalka D (1992) Phar-
macokinetic and metabolic aspects of the moclobemide-food interaction. Psycho-
pharmacology 106:S37-S39
Cole SK, Story MJ, Laudanski T, Dwyer M, Attwood D, Robertson J, Barnwell SG
(1992) Targeting drugs to the enterohepatic circulation: a potential drug delivery
system designed to enhance the bioavailability of indomethacin. Int J Pharm
80:63-73
Confalonieri S, Cosmi G, Guiso G, Gherardi S, Guido M, Caccia S (1992) The pharma-
cokinetics of a potential memory-enhancing compound, CL 275838, in fasted and
fed volunteers. Drug Invest 4:322-328
Conway EL, McNeil JJ, Hurley J, Jackman GP, Krum H, Howes LG, Louis WJ (1993)
The effects of food on the oral bioavailability of doxazosin in hypertensive sub-
jects. Drug Invest 6:90-95
Cook CS, Hauswald CL, Grahn A Y, Kowalski K, Karim A, Koch R, Schoenhard GL,
Oppermann JA (1990) Suitability of the dog as an animal model for evaluating
theophylline absorption and food effects from different formulations. Int J Pharm
60:125-132
Cook HJ, Mundo CR, Fonseca L, Gasque L, Moreno-Esparza R (1993) Influence of
the diet on bioavailability of tetracycline. Biopharm Drug Dispos 14:549-553
De Mey C, Meineke I (1992) Prandial and diurnal effects on the absorption of orally
administered enteric coated 5-aminosalicylic acid (5-ASA). Br J Clin Pharmacol
33:179-182
Degen PH, Cardot JM, Czendlik C, Dieterle W (1993) Influence of food on the
disposition of the monoamine oxidase-A inhibitor brofaromine in healthy volun-
teers. Biopharm Drug Dispos 14:209-215
Degen PH, Flesch G, Cardot J-M, Czendlik C, Dieterle W (1994) The influence of food
on the disposition of the anti epileptic oxcarbazepine and its major metabolites in
healthy volunteers. Biopharm Drug Dispos 15:519-526
Desante KA, Zeckel ML (1992) Pharmacokinetic profile of loracarbef. Am J Med
92:16S-19S
Desmond PV, Harman PJ, Gannoulis N, Kamm M, Mashford ML (1990) The effect of
an antacid and food on the absorption of cimetidine and ranitidine. J Pharm
Pharmacol 42:352-354
Devane JG, Kelly JG (1991) Effect of food on the bioavailability of a multiparticulate
sustained-release verapamil formulation. Adv Ther 8:48-53
Devane JG, Kelly JG, Geoghegan B (1990) Pharmacokinetic and in vitro
characteristics of sustained release verapamil products. Drug Dev Ind Pharm
16:1233-1248
DeVries TM, Voigtman RE, Posvar EL, Nesbitt RU, Forgue ST (1991) Pharmacoki-
netic characterization of a procainamide tablet suitable for twice daily dosing.
Pharm Res 8 [Suppl]:S315
Doose DR, Gisclon LG, Stellar SM, Riffits JM, Hills JF (1992a) The effect of food on
the bioavailability of topiramate from 100- and 400-mg tablets in healthy male
subjects. Epilepsia 33 [Suppl 3]:105
Doose DR, Minn FL, Stellar S, Nayak RK (1992b) Effects of meals and meal compo-
sition on the bioavailability of fenretidine. J Clin Pharmaco132:1089-1095
Dressman JB, Berardi RR, Elta GH, Gray TM, Montgomery P A, Lau HS, Pelekoudas
KL, Szpunar GL, Wagner JG (1992) Absorption of flurbiprofen in the fed and
fasted states. Pharm Res 9:901-907
Drew RH, Gallis HA (1992) Azithromycin-spectrum of activity, pharmacokinetics, and
clinical applications. Pharmacotherapy 12:161-173
114 P.G. WELLING
Gupta SK, Benet LZ (1990) High-fat meals increase the clearance of cyclosporine.
Pharm Res 7:46-48
Gupta SK, Manfro RC, Tomlanovich SJ, Gambertoglio JG, Garovoy MR, Benet LZ
(1990) Effect of food on the pharmacokinetics of cyclosporine in healthy subjects
following oral and intravenous administration. J Clin Pharmacol 30:643-653
Haegele KD, Hinze C, Jode-Ohlenbusch A-M, Creamer G, Borlack J (1991) Effects of
a standardized meal on the pharmacokinetics of the new cardiotonic agent
piroimone. Arzneimittelforschung 41:1225-1229
Hamaguchi T, Shinkuma D, Irie T, Yamanaka Y, Morita Y, Iwamoto B, Miyoshi K,
Mizuno N (1993) Effect of a high-fat meal on the bioavailability of phenytoin in a
commercial powder with a large particle size. Int J Clin Pharmacol 31:326-330
Hanyok 11 (1993) Clinical pharmacokinetics of sotalol. Am J CardioI12:19A-26A
Harrison LI, Riedel DJ, Armstrong KE, Goldlust MB, Ekholm BP (1992) Effect of
food on salsalate absorption. Ther Drug Monit 14:87-91
Harrison LI, Mitra AK, Kehe CR, Klinger NM, Wick KA, McCarville SE, Cooper KM,
Chang SF, Roddy PJ, Berge SM, Kisicki JC, Dockhorn R (1993) Kinetics of
absorption of a new once-a-day formulation of theophylline in the presence and
absence of food. J Pharm Sci 82:644-648
Hilleman DE, Mohiuddin SM, Destache CJ, Stoysich AM, Nipper HC, Maleskar MA
(1992) Impact of food on the bioavailability of encainide. J Clin PharmacoI32:833-
837
Hoke JF, Chi EM, Anthony KK, Kulmala HK, Sussman NM, Okerholm RA (1991)
Effect of food on the bioavailability of vigabatrin tablets. Epilepsia 32 [SuppI3]:6-
7
Holmes B, Brogden RN, Richards DM (1985) Norfioxacin: a review of its antimicrobial
activity, pharmacokinetic properties and therapeutic use. Drugs 30:482-513
Honcharik N, Yatscoff RW, Jeffery JR, Rush DN (1991) The effect of meal composi-
tion on cyclosporine absorption. Transplantation 52:1087-1089
Honma A, Ikeda K, Udo N, Sdamori M, Hasegawa K, Kuruma I, Uto M, Nakahara T,
Fujita T (1986) Aniracetam: clinical phase I study of aniracetam. J Clin Ther Med
2:929-952
Hoon TJ, McCollam PL, Beckman KJ, Hariman RJ, Bauman JL (1992) Impact of food
on the pharmacokinetics and electrocardiographic effects of sustained release
verapamil in normal sUbjects. Am J Cardiol 70:1072-1076
Hooper WD, Dickinson RG, Eadie MJ (1990) Effect of food on absorption of
lomefioxacin. Antimicrob Agents Chemother 34:1797-1799
Hopkins A (1966) The pattern of gastric emptying: a new review of old results. J
Physiol (Lond) 182:144-149
Hughes WT, Kennedy W, Shenep JL, Flynn PM, Hetherington SV, Fullen G,
Lancaster DJ, Stein DS, Palte S, Rosenbaum D, Lian SHT, Blum MR, Rugers M
(1991) Safety and pharmacokinetics of 566C80, a hydroxynaphthoquinone with
anti-Pneumocystis carinii activity: a phase I study in human immunodeficiency
virus (HIV)-infected man. J Infect Dis 163:843-848
Hunt TL, Kaiko RF (1991) Comparison of the pharmacokinetic profiles of two oral
controlled-release morphine formulations in healthy young adults. Clin Ther
13:482-488
Hussey EK, Donn KH, Powell JR, Lahey AP, Pakes GE (1991) Albuterol extended-
release products: effect of food on the pharmacokinetics of single oral doses of
Volmax® and Proventil® repetabs in healthy male volunteers. J Clin Pharmacol
31:561-564
Hyneck MA (1992) An overview of the clinical pharmacokinetics of nabumetone. J
Rheumatol 19 [Suppl 36]:20-24
Ingwersen SH, Mant TGK, Larson 11 (1993) Food intake increases the relative oral
bioavailability of vanoxerine. Br J Clin PharmacoI35:308-310
Iseki K, Satoh Y, Sugawara M, Miyazaki K (1994) Effect of Enterued® administration
on the intestinal absorption of orally active cefem antibiotics. J Pharm Soc Jpn
114:233-240
116 P.G. WELLING
Jallad NS, Callejas HJ, Woo-Ming RB, Weidler DJ (1991) The pharmacokinetics of
astemizole plus pseudoephedrine when taken with a standardized meal. J Clin
Pharmacol31:863
Jarvinen A, Nykanen S, Mattila J, Haataja H (1992) Effect of food on absorption and
hydrolysis of erythromycin acistrate. Arzneimittelforschung 42:73-76
Johnson BF, O'Grady J, Sabey GA, Bye C (1978) Effect of a standard breakfast on
digoxin absorption in normal subjects. Clin Pharmacol Ther 23:315-319
Kaiko R, Grandy R, Thomas G, Goldenheim P (1990) A single-dose study of the effect
of food ingestion and timing of dose administration on the pharmacokinetic profile
of 30mg sustained-release morphine sulfate tablets. Curr Ther Res 47:869-878
Kann J, Levitt MJ, Horodniak JW, Pay JW (1989) Food effects on the nighttime
pharmacokinetics of Theo-Dur tablets. Ann Allergy 63:282-286
Keown PA, Stiller CR, Laupacis AL, Howson W, Coles R, Stawecki M, Koegler J,
Carruthers G, McKenzie N, Sinclair NR (1982) The effects and side effects of
cyclosporine: relationships to drug pharmacokinetics. Transplant Proc 14:659-661
KivistO KT, Ojala-Karlsson P, Neuvonen PJ (1992) Inhibition of norfloxacin absorp-
tion by dairy products. Antimicrob Agents Chemother 36:489-491
Klamerus KJ, Troy SM, Ben-Maimon CS, Chiang ST (1993) Effect of age, gender, and
food on zalospirone disposition. Clin Pharmacol Ther 53:193
Kleinbloesem CH, Ouwerkerk M, Brodenfeldt R (1993) Food effect with extended
release formulations: nifedipine. Clin Pharmacol Ther 53:207
Knupp CA, Milbrath R, Barbhaiya RH (1993) Effect of time of food administration on
the bioavailability of didanosine from a chewable tablet formulation. J Clin
Pharmacol 33:568-573
Kopitar Z, Vrhovac B, Povsic L, Plavsic F, Francetic I, Urbancic J (1991) The effect of
food and metoclopramide on the pharmacokinetics and side effects of
bromocriptine. Eur J Drug Metab Pharmacokinet 16:177-181
Korth-Bradey JM, Fruncillo RJ, Klamerus K-J, Chiang ST, Conrad KA (1992) The
influence of high and low fat meals on single dose pharmacokinetics of
zalospirone. Pharm Res 9 [Suppl]:S326
Kosoglou T, Kazierad D, Schentag JJ, Patrick JE, Heimark L, Flannery BE, Affrime
MB (1993) Effect of food and gastric emptying rate (GER) on the bioavailability
(BA) of 5-ISMN. Clin Pharm Ther 53:206
Kozloski GD, de Vito JM, Johnson JB, Holmes GB, Adams MA, Hunt TL (1992a)
Bioequivalence of verapamil hydrochloride extended-release pellet-filled capsules
when opened and sprinkled on food and when swallowed intact. Clin Pharm
11:539-542
Kozloski GD, De Vito JM, Kisicki JC, Johnson JB (1992b) The effect of food on the
absorption of methotrexate sodium tablets in healthy volunteers. Arthritis Rheum
35:761-764
Kuhn M (1993) Drug interactions and their nursing implications. J NY State Nurses
Assoc 24:10-16
Lafontaine D, Mailhot C, Vermeulen M, Bissonnette B, Lambert C (1990) Influence of
chewable sucraflate or a standard meal on the bioavailability of naproxen. Clin
Pharm 9:773-777
Lasswell AB, Loreck ES (1992) Development of a program in accord with JACAHO
standards for counseling on potential drug-food interactions. J Am Diet Assoc
92: 1124-1125
Latini R, Tognoni G, Kates RE (1984) Clinical pharmacokinetics of amiodarone. Clin
Pharmacokinet 9:136-156
Lecaillon JB, Dubois JP, Soula G, Pichard E, Poltera AA, Ginger CD (1990) The
influence of food on the pharmacokinetics of CGP 6140 (amocarzine) after oral
administration of a 1200mg single dose to patients with onchocerciasis. Br J Clin
Pharmacol 30:629-633
Lecaillon JB, Poltera AA, Zea-Flores G, De Ramirez I, Nowell De Arevalo A (1991)
Influence of food related to dose on the pharmacokinetics of amocarzine and of its
Drug-Food Interactions Affecting Drug Absorption 117
Pan HY, Devault AR, Brescia D, Willard DA, McGovern ME, Whigan DB, Ivashkiv
E (1993) Effect of food on pravastatin pharmacokinetics and pharmacodynamics.
Int J Clin PharmacoI31:291-294
Palazzini E, Cristoformi M, Babbini M (1992) Bioavailability of a new controlled-
release oral naproxen formulation with and without food. Int J Clin Pharmacal
Res 12:179-184
Peck RW, Weatherley BC, Posner J (1993) The pharmacokientics of 882C, a thymidine
analogue with potent anti-VZV activity in healthy volunteers. Antiviral Res 20
[Suppl 1]:132
Pfeifer S (1993) EinfluB von Nahrung auf die Pharmakokinetik von Arzneistoffen.
Pharmazie 48:3-16
Pfeiffer A, Vidon N, Bovet M, Rongier M, Bernier 11 (1990) Intestinal absorption of
amiodarone in man. J Clin Pharmacol 30:615-620
Phelan MJI, Orme M, Williams E, Thompson RN (1991) Food has no effect on the
pharmacokinetics of methotrexate in patients with rheumatoid arthritis. Arthritis
Rheum 34 [Suppl]:S91
Pieniaszek HJ, Rakestraw DC, Schary WL, William RL (1991) Influence of food on the
oral absorption and bioavailability of moricizine. J Clin PharmacoI31:792-795
Plezia PM, Thornley SM, Kramer TH, Armstrong EP (1990) The influence of enteral
feedings on sustained-release theophylline absorption. Pharmacotherapy 10:356-
361
Plusquellec Y, Compistron G, Stevens S, Barre J, Jung J, Tillement JP, Houin G (1987)
A double-peak phenomenon in the pharmacokinetics of veralipride after oral
administration: a double site model for drug absorption. J Pharmacokinet
Biopharm 15:225-239
Prescott LF, Yoovathaworn K, Makarananda K, Saivises R, Sriwatanakul K (1993)
Impaired absorption of paracetamol in vegetarians. Br J Clin Pharmacol 36:237-
240
Ptachcinski RJ, Venkataramanan R, Rosenthal JT, Burckart GJ, Taylor RJ, Hakala
TR (1985) The effect of food on cyclosporine absorption. Transplantation 40:174-
176
Rapin J-R (1992) Effect of food on calcium antagonists pharmacokinetics. Pharm
Weekbl14 [Suppl E]:E5
Riva E, Gerna M, Latini R, Giani P, Volpi A, Moggioni A (1982) Pharmacokinetics of
amiodarone in man. J Cardiovasc Pharmacol 4:264-269
Roberts RJ, Leff RD (1989) Influence of absorbable and non absorbable lipids and
lipidlike substances on drug bioavailability. Clin Pharmacol 45:299-304
Robertson DRC, Higginson I, MacKlin BS, Renwick AG, Waller DG, George CF
(1991) The influence of protein containing meals on the pharmacokinetics of
levadopa in healthy volunteers. Br J Clin PharmacoI31:413-417
Roe AR (1989) Diet and drug interactions. Van Nostrand Reinhold, New York
Rolan PE, Mercer AJ, Westherley BC, Holdich T, Ridout G, Meire H, Posner J (1992)
Investigation of the factors responsible for a food-induced increase in absorption
of a novel antiprotozoal drug 566C80. Br J Clin Pharmacol 33:226P-227P
Rolan PE, Mercer AJ, Weatherley BC, Holdich T, Meire H, Peck RW, Ridout G,
Posner J (1994) Examination of some factors responsible for food-induced in-
crease in absorption of atovaquone. Br J Clin Pharmacol 37:13-20
Roller S, Lode H, Stelzer I, Deppermann KM, Boeckh M, Koeppe P (1992) Pharmaco-
kinetics of loracarbef and interaction with acetylcysteine. Eur J Clin Microbiol
Infect Dis 11:851-855
Roncari G (1993) Human pharmacokinetics of aniracetam. Drug Invest 5 [Suppl1 ]:68-
72
Roos RAC, Tijssen MAJ, Van der Velde EA, Breimer DD (1993) The influence of a
standard meal on Sinemet CR absorption in patients with Parkinson's disease.
Clin Neurol Surg 95:215-219
Rosenborg J, Nyberg L, Delen A-M (1991) Dietary habits do not influence the dosage
of Bambec® tablets. Eur Respir J 4 [Suppl 14]:434S
120 P.G. WELLING
Yogendran L, McLaughlin K, Vadas EB, Margolskee DJ, Bechard S, Hseih JY-K, Lin
C, Rogers JD, Sciberras DG, James 1M (1992) Comparative bioavailability of
different formulations of MK-679 an orally active LTD4 receptor antagonist. Br J
Clin PharmacoI34:169P-170P
Yong C-L, Yu D, Eden L, Eichmeyer L, Giessing D (1991) Effect of food on the
pharmacokinetics of oxybutinin in normal subjects. Pharm Res 8 [Suppl]:S320
Yu DK, Morrill B, Bhargava VO, Giesing DH, Weir SJ (1992) Effect of food
coadministration on Cardizem CDTM capsule bioavailability. Pharm Res 9 [Suppl]:
S323
Yuen KH, Deshmukh AA, Newton JM, Short M, Melchor R (1993) Gastrointestinal
transit and absorption of theophylline from a multiparticulate controlled release
formulation. Int J Pharm 97:61-77
Zimmermann T, Yeates RA, Laufen H, Pfaff G, Wildfeuer A (1994) Influence of
concomitant food intake on the oral absorption of two triazole antifungal agents,
itraconazole and fluconazole. Eur J Clin PharmacoI46:147-150
CHAPTER 4
Drug Interactions at Plasma
and Tissue Binding Sites
J.e. McELNAY
A. Introduction
The binding of drugs to plasma and tissue proteins is a major determinant of
drug distribution throughout the body. This binding has also a very important
effect on drug dynamics since it is only the free (unbound) drug which can
diffuse to, and interact with, receptor sites, i.e., bound drug is pharmacologi-
cally inactive. The relative ease of study in vitro of the binding of drugs in
human plasma and in separate protein fractions (mainly albumin) has led to an
extensive literature on the subject, with the influence of other drugs and/or
endogenous materials on drug binding being a common topic for investigation.
The relative difficulty in obtaining samples of viable human tissues has meant
that work on the binding of drugs to human tissues has been very limited by
comparison, although this may change with the more widespread use of tissue
culture techniques. It is as a result of this large body of largely in vitro data that
the topic of protein binding displacement interactions has gained prominence
as a possible mechanism of drug-drug interactions; however, as mentioned in
Chap. 1 of this volume, the importance of plasma protein binding displace-
ment interactions has been overestimated and overstated and only in very
specific cases do they result in adverse outcomes. In this chapter the influence
of plasma and tissue binding on drug kinetics is discussed to facilitate consid-
eration of the consequences of drug displacement from binding sites and their
possible clinical significance. Those cases in which displacement can lead to
clinically significant outcomes, which are much more restrictive than much of
the literature and popular belief amongst practitioners would suggest, are
considered in some detail. Before moving on the these issues, the proteins
involved in drug binding are described.
... .
.
...
compartment, drugs which are highly bound to plasma proteins tend to have
low apparent volumes of distribution (Vd). Warfarin, the best-known example
of a drug which is highly bound to plasma albumin (>99%), has a low Vd (0.111
kg), indicating poor diffusion into the tissues (TILLEMENT et al. 1978). Drugs
which are highly bound to tissues tend to have a high Vd. This, of course, will
depend on the type of tissue to which the drug is bound and its abundance in
the body. For drugs which are highly bound to both plasma and tissues, the Vd
will depend on the relative binding in both sites. With tricyclic antidepressants
and phenothaizines, for example, almost all of the drug in plasma is bound to
albumin; however, due to extensive tissue binding of these agents, the circulat-
ing drug in plasma represents only a small fraction of the total drug in the body
(KOCH-WESER and SELLERS 1976). The equilibrium which exists between
tissue and plasma binding is represented in Fig. 2. As mentioned earlier, only
unbound drug is available to diffuse to receptor sites and exert therapeutic
effects.
The next phase in the pharmacokinetic process is drug elimination, involv-
ing primarily the liver and kidney. It is obvious that if a drug is highly bound
to tissues, it will be protected against elimination since much of the drug
will be outside the plasma compartment and will therefore not be "delivered"
to the kidney or liver for elimination. In addition, the extent of plasma
protein binding can have marked effects on drug clearance by these organs.
Consider first drug metabolism. For most drugs, plasma protein binding is
protective in that the affinity for plasma binding sites exceeds that for
metabolising systems. It is therefore only free (unbound) drug that is
metabolised during passage through the liver (or other metabolic site). Such
drugs would be considered to have a low hepatic extraction ratio (low
tendency to be removed from the blood during its passage through the liver).
On the other hand, if a drug has a high extraction ratio, the attraction to
metabolic enzyme systems in the liver will overcome the binding affinity for
plasma proteins and bound drug will be stripped from the binding sites to
undergo metabolic change (McELNAY and D'ARCY 1983). This will often
involve active uptake of the drug into liver cells. In the case of high extraction
drugs, plasma protein binding will facilitate drug metabolism by maintaining
high total drug concentrations in the plasma (via decreased drug distribution)
and will essentially act as a carrier system, taking the drug to its site of
destruction. The metabolism of highly extracted drugs, e.g., propranolol, are
dependent upon liver blood-flow (ROWLAND 1980).
A similar situation pertains to the kidney. Glomerular filtration, being
a passive process, allows only free (unbound) drug to pass through the
glomerulus in the normal healthy kidney, since protein molecules are too
large to be filtered. Plasma protein binding therefore protects a drug against
elimination in the kidney by glomerular filtration. As the filtrate passes down
the renal tubules, back diffusion of drug occurs when the concentration of
free drug in the renal tubule (due to water reabsorption) exceeds the free
Drug Interactions at Plasma and Tissue Binding Sites 129
Plasma Tissues
Bound Bound
drug drug
II
Free .c::.
1l
Free
drug drug
/'
Fig.2. Distribution of drug between plasma and tissue compartments. Only free (phar-
macologically active) drug can diffuse out of the plasma; the equilibrium would there-
fore move towards the right after displacement from plasma binding and towards the
left after displacement from tissue binding sites. Free drug concentrations in the plasma
and tissue compartments are equal. Quantities of drug bound will depend on the
amount of binding materials present in both plasma and tissues and their respective
binding capacities. (After TILLEMENT 1980)
from its binding. In this type of interaction the phenylbutazone is termed the
"displacer" drug and warfarin the "displaced" drug, the latter term being
usually reserved for the drug with the higher potential to give rise to adverse
effects (KOCH-WESER and SELLERS 1976). Since the displacement is competi-
tive, the binding of both drugs will be decreased. For substantial displacement
to take place, the displacer must occupy the majority of the available binding
sites, thereby substantially lowering the binding sites available to the primary
drug (ROWLAND and TOZER 1995). For this to take place the molar concentra-
tion of drug in plasma must exceed the molar concentration of albumin
(approximately 150.ug/ml for a drug with a molecular weight of 250). As a
result of these prerequisites relatively few drugs are commonly referred to as
displacers, e.g., salicylic acid, valproic acid and phenylbutazone. Since the
molar concentration of lXt-acid glycoprotein is 9-23.uM in plasma, lower molar
concentrations of basic drugs will be required to fulfil the latter displacer
criterion (ROWLAND and TOZER 1995). In certain cases the parent drug may not
itself be involved in a displacement interaction, but rather a metabolite may
act as the displacer, e.g., trichloracetic acid displaces warfarin while the parent
drug chloral hydrate does not (SELLERS and KOCH-WESER 1970).
As well as the competitive displacement interactions, the binding of one
drug to, e.g., albumin, can give rise to changes in the conformation of the
protein and thereby change the shape of the binding sites for a second drug.
This normally gives rise to a decreased binding of the second drug although
occasionally with allosteric effects the binding affinity of the second drug is
increased. Aspirin influences the binding of certain drugs by this non-competi-
tive mechanism, e.g., flufenamic acid and phenylbutazone, via permanently
acetylating lysine residues of albumin (PINKARD et al. 1973). The alkylating
agents carmustine and mechlorethamine give rise to decreased binding of
penbutolol to lXt -acid glycoprotein. Dialysis against saline for 24 h does not
restore the free fraction of penbutolol. The authors who conducted this latter
research concluded that treatment of cancer patients with alkylating agents
could alter the serum proteins and modify their binding capacity and that this
should be taken into account when co-administering other basic drugs, e.g.,
methadone (AGUIRRE et al. 1994).
The study of the binding of drugs to tissue components had been much
more limitied that those involving plasma; however, it is likely that both
competitive and non-competitive binding displacement also takes place at
tissue binding sites.
E. Therapeutic Consequences
of Plasma Binding Displacement Drug-Drug Interactions
The interest in plasma protein binding displacement interactions was first
aroused by the well-known interaction between warfarin and phenylbutazone
(AGGELER et al. 1967), which results in a marked, and clinically significant,
Drug Interactions at Plasma and Tissue Binding Sites 131
c:
Displacing
.~
~
Agent
2
C
cu
u
c:
o
U
I
o
Iii L..
..2
a..
o
....o
e
0>
o
.....J
TIME
Fig. 3. Typical plasma concentration-time profile for a drug interaction involving
plasma protein binding placement. An almost immediate drop in total plasma concen-
tration (due to an increased Vd) is seen. Assuming clearance is unchanged, elimination
half-life in this case is shown to be increased (decreased slope of elimination curve).
(After D'ARCY and McELNAY 1982)
were increased as was the Vd, while plasma protein binding was decreased
(BECK et al. 1990).
If follows therefore that interactions which have in the past been attrib-
uted solely to plasma binding displacement must have a different underlying
mechanism. The best example of this is the now familiar warfarin-
phenylbutazone interaction. As long ago as 1974, LEWIS et al. discovered a
metabolic mechanism for this interaction. Phenylbutazone was shown to ste-
reo-selectively inhibit the metabolism of S-warfarin (warfarin is available
commercially as a racemic mixture and S-warfarin is five to six times more
potent than R-warfarin; O'REILLY 1971). There was a corresponding increase
in the elimination of R-warfarin. The increased proportion of the more potent
S-warfarin, however, gave rise to the increased anticoagulation observed.
Azapropazone is another example of a non-steroidal anti-inflammatory agent
which gives rise to markedly increased prothrombin times when administered
together with warfarin (POWELL-JACKSON 1977). Although azapropazone is a
potent displacer of warfarin from plasma binding sites (McELNAY and D'ARCY
1980), this displacement cannot explain the increased pharmacological
effect seen, and, although it has never been investigated, it would appear
that a metabolic effect similar to that with phenylbutazone takes place
(azapropazone has structural similarities to phenylbutazone). (Although many
other non-steroidal anti-inflammatory agents may not increase prothrombin
time, it should be borne in mind that they can cause gastrointestinal bleeding,
which can be exacerabated in anticoagulated patients.)
Drug Interactions at Plasma and Tissue Binding Sites 133
F. Therapeutic Consequences
of Tissue Binding Displacement Interactions
Displacement of drugs from tissue binding sites gives rise to the opposite effect
to that of plasma binding displacement, i.e. the equilibrium depicted in Fig. 2
moves in a clockwise direction, giving rise to redistribution of drug from the
tissues back into the plasma compartment. The plasma compartment, being
relatively small in comparison to the tissue compartment, may be unable to
buffer the impact of increasing amounts of free drug and therefore adverse
sequelae may be more likely. Since increased free drug is forced into the
plasma compartment, the total (free + bound) drug concentration will initially
be increased (Fig. 4) and there will be a reduction in the apparent volume of
distribution of the displaced drug. Elimination mechanisms will of course
come into play and the rate of elimination of the displaced drug in, for
example, the liver and the kidney will increase due to increased concentrations
of free drug. Assuming binding within the plasma remains unaffected and drug
clearance remains the same, both the mean total and the mean free drug
concentrations will re-equilibrate to their original pre-interaction values. This
re-equilibration, unlike the case with plasma binding displacement in which
redistribution into the "sink" of the tissues takes place very quickly, will
require some four to five half-lives. Therefore for some drugs with a long
half-life, mean plasma concentrations of free drug may remain elevated for
prolonged periods. Although clearance will not be effected, the reduction
in Vd will result in a decrease in the elimination half-life, which in turn will
lead to greater than normal fluctuations in plasma levels of the displaced
drug during a dosing interval. A major drug interaction can take place if
the displacing drug also inhibits the elimination of the displaced agent as
occurs in some of the known interactions which occur by this mechanism. The
best-known interactions that involve tissue displacement are with the cardiac
glycoside, digoxin. The most extensively studied interaction involves digoxin
and quinidine (Fig. 5). Administration of quinidine to a patient stabilised on
digoxin leads, as expected, to an increased digoxin concentration in plasma. In
one clinical trial, serum digoxin concentrations increased in 25 of 27 subjects
during co-administration of quinidine from a mean of 1.4ng/ml to 3.2ng/ml.
Signs of digoxin toxicity (anorexia, nausea and vomiting) were noted in 16
patients (59%) during concomitant quinidine therapy (REIFFEL et al. 1979).
The data presented in Fig. 5 indicate clearly that the volume of distribution
of digoxin is decreased during concomitant quinidine treatment - mean
Drug Interactions at Plasma and Tissue Binding Sites 137
c
.2
~c
II)
u
c
0
t
u
0
~
0
a: D isplocing
0 Agent
"0
~
0
OJ
0
-'
TIME
Fig.4. Typical plasma concentration time profile for a drug interaction involving tissue
binding displacement. An almost immediate increase in total plasma concentration
(due to a decreased Vd) is seen. Elimination half-life is shown to be increased. This is
because of increased amounts of both free and bound drug being presented at sites of
elimination. (After D' ARCY and McELNAY 1982)
40.0
30.0
20.0
10.0
5.0
10 20 30 40 50 60 70 80 90
Time (hours)
values for Vd were 1O.871Jkg for digoxin alone and 7.351/kg for digoxin
administration to six patients receiving quinidine (HAGER et al. 1979). These
authors suggested that the results could be explained by the displacement
of digoxin from tissue binding sites by quinidine. Renal clearance of digoxin
is also decreased by quinidine, probably due to inhibition of renal tubular
secretion, and therefore this compensatory mechanism is compromised, a
situation which will further exacerbate the outcome of the interaction. It is
obviously necessary to carefully monitor serum digoxin concentrations during
concomitant quinidine treatment and, indeed, DOERING (1979) has suggested
that digoxin dosage should be halved during quinidine therapy, as on average
digoxin concentration doubles during the interaction. There is, however, no
substitute to concentration monitoring since there is great variability in the
magnitude of the quinidine-induced effects, e.g., one patient may have no
induced increase in digoxin serum concentration while another may have a
fivefold increase (BIGGER 1979). Since discovery of the interaction involving
quinidine, several other structurally related drugs have been shown to be
involved in a similar interaction with digoxin. These drugs include the other
antiarrhythmics amiodarone and propafenane. Disopyramide is free from
interaction with digoxin and could be used to replace quinidine in patients
receiving digoxin, since it has similar properties and indications (KOCH-WESER
1979). Choroquine, an antimalarial drug, which is also used as a disease
modifying agent in the treatment of rheumatoid arthritis, has been shown to
increase digoxin plasma levels in dogs, presumably via a tissue binding
displacement interaction (McELNAY et al. 1985).
An interaction involving both displacement at plasma and tissue binding
sites will have an even greater potential for adverse effects since free drug
concentrations could be elevated to an even more marked extent.
Sulphaethidole, for example, may displace dicloxacillin at both sites (DE
SANTE et al. 1980).
with heparin to keep them patent during clinical trials, such an effect
could give rise to erroneous data. Heparin, for example, has also been shown
to alter the plasma binding of phenytoin (CRAIG et al. 1976), quinidine
(KESSLER et al. 1979), propranolol (WOOD et al. 1979a,b) and warfarin
(CHAKRABARTI 1978).
Although initially the displacement via endogenous fatty acids appeared
to be an in vivo effect, a careful study by GIACOMINI et al. (1980) indicated that
the displacement takes place after sample collection. Via in vitro hydrolysis of
plasma triglycerides by lipases, which are released and activated after in vivo
administration of heparin, free fatty acid concentrations in blood samples are
increased. The problem can largely be overcome by the use of a lipase inhibi-
tion in the sample collection vial and it is recommended that this precaution be
taken when performing protein binding analyses with drugs which can be
displaced by free fatty acids.
40
E % Free 100M
35
... % FreeCtri
E
30
Q)
25
~
~
0
:Q
u 20
ro
u
'0
0..
<ii
>
15
,... ...
... ...
10
...
5
20 25 30 35 40 45 50 55
Serum albumin concentration (giL)
1978; 0IE 1986). All these conditions will complicate drug-drug interactions at
plasma binding sites. DASGUPTA et al. (1994) found that endogenous com-
pounds present in uraemic serum led to a decreased binding of valproate, but
that the displacement of valproate by salicylate was less marked in uraemic
serum when compared with normal serum. A review of the influence of disease
on plasma protein binding has been presented by GRAINGER-RoUSSEAU et al.
(1989) and a summary table from that review is presented as Table 1. In the
present context, this table serves two purposes; first it draws attention to
specific drug-disease interactions involving plasma protein binding and sec-
ondly data on the free fraction of a range of drugs which are bound to plasma
proteins are presented. The latter information is important bearing in mind
that clinically significant drug-drug displacement interactions in plasma usu-
ally involve drugs which are greater than 90% bound to plasma proteins at
normal therapeutic concentrations. Although pregnancy is not a disease, it has
been included in Table 1 since binding of some drugs will change during
pregnancy. It has recently been suggested, for example, that during pregnancy
and the 1st month after delivery, both total and free valproate serum concen-
trations should be closely monitored to determine the lowest effective dose
(JOHANNESSEN 1992).
Drug Interactions at Plasma and Tissue Binding Sites 141
Carbamazepine Alcoholic liver 22.7% vs. 19.5% Y Not clinically significant. BARRUZI
disease, renal failure, Caution in interpreting et a1. 1986
rheumatoid arthritis, total drug concentrations.
ulcerative colitis Correlation with AGP
shown
Diazepam Early pregnancy 1.8% vs. 1.9% N Large V d and therapeutic PERUCCA
Mid pregnancy 1.8% vs. 2.1 % Y index means change is not et a!. 1981
Late pregnancy 1.8% vs. 2.6% Y clinically important in
chronic use. If used in
status epilepticus may have
potentiated action in late
pregnancy due to increased
free fraction
Uraemia 2.9% vs. 4.5% Not indicated. Site II not CALVO
affected by carbamylation. et al. 1982
Endogenous binding
inhibitors implicated
Uraemia 1.64 % vs. 3.23 % Y Not indicated, but an GROSSMAN
Kidney transplant 1.50% vs. 2.11 % Y important difference et a!. 1982
Nephrotic syndrome 1.60% vs. 3.55% Y implicated when
interpreting kinetic data
with regard to type of renal
renal disease and the
protein involved in binding
Chronic cardiac "=1 % VS. 1.5% N Does not alter diazepam FiTCHL
failure binding site affinity et al. 1983
Acute uraemia 2% VS. 6% Y Large therapeutic index TJULA and
Chronic uraemia 2% vs. 4% Y means that drug effect can NEUVONEN
be monitored clinically as 1986
clinical end point can be
determined safely
Age-related decrease 1.8% vs. 3.0% Y Pharmacokinetic changes TIULA and
in renal function but no clinical implication ELFVING
1987
Digitoxin Chronic cardiac 6% vs. 5.9% N Not significant FITCHL
failure et a1. 1983
End-stage renal 2.0% vs. 2.5% N Difference too small to LOHMAN
disease have any therapeutic and
(haemodialysis) consequence MERKUS
1978
Disopyramide MI 0.25%, vs. 0.15% Y Binding varies with drug DAVID
(at 2mgll) and protein concentration. et a1. 1983
0.53% vs. 0.32% May be clinically significant
(at 5mgll) as AGP concentration
decreases post MI
MI 0.2% vs. 0.13% Y Not indicated. CAPLIN
Pharmacokinetic change et a!. 1985
suggested. Interpretation of
total drug levels caution
Flecainide MI 39% vs. 47% Y Not indicated. Endogenous CAPLIN
compounds may lead to et a!. 1985
displacement and
decreased binding
Imipramine Chronic cardiac 19% vs. 18% N Drug binding not affected FrrcHL
failure et a1. 1983
Lignocaine Uraemia 30.7% vs. 20.8% Y Interpreting kinetic data GROSSMAN
142 J.e. McELNAY
Table 1. Continued
Drug Disease % Free or free Significant Clinical significance! References
fraction (control difference comment
vs. patient)
Kidney transplant 33.7% vs. 24.6% Y with regard to the type of et al. 1982
renal disease and the
Nephrotic syndrome 30.4 % vs. 34.2 % N protein involved in
binding may be important
NIDDM 32% vs. 30% N Not indicated (but no O'BYRNE
change expected) et al. 1988
Lorcainide Cardiac arrhythmia 26.03% vs. 24.07% N Not significant SOMANI
et al. 1984
Renal disease 26.03% vs. 29.04% N Not significant
Metociopramide Renal disease 0.6% vs. 0.59% N Not significant WEBB et al.
1986
Phenylbutazone Alcoholic hepatitis 6% vs. 13% Y Not known. BRODIE
Alcoholic cirrhosis 6% vs. 19% Y Bilirubin and and BOOBIS
hypoalbuminaemia 1978
implicated in binding
defect
Phenytoin Early pregnancy' 9.7% vs. 10.6% Y Important in drug PERUCCA
Mid pregnancy 9.7% vs. 10.9% Y monitoring interpretation. et al. 1981
Late pregnancy 9.7% vs. 12.6% Y Decreased albumin
concentration in
pregnancy implicated
Chronic cardiac = 15% vs. 15% N Not significant FITCHL
failure et al. 1983
Acute uraemia 10% vs. 25% Y Important clinically in drug TIULA and
Chronic uraemia 10% vs. 24% Y monitoring, although free NEUVONEN
concentration remains the 1986
same due to
pharmacokinetic
compensation
Age-related 10% vs. 13.5% Y Important in drug TIULA and
decrease in monitoring ELFVlNG
renal function 1987
Prednisolone Portosystemic 17.7% vs. 28.6% Y Not significant as BERGREM
shunt elimination and V d altered; et al.1983
therefore, normalising free
concentration
Nephrotic syndrome 2.26 x l(J3vs. Y Not known. Altered FREY and
4.20 x 103 M-I pharmacokinetic FREY 1984
(albumin K,) disposition, due to
2.12 x 10' vs. N change in binding
3.44 x 10' M-I
(transcortin K,)
Propranolol Chronic cardiac 14% vs. 14% N AGP binding not affected FITCHL
failure et al. 1983
Cancer, 10.8% vs. 5.5% Y Not clinically significant PAXTON
MI and IHD, but may be important when and BRIANT
infection, heart interpreting drug levels 1984
failure, COPD,
CVA, miscellaneous
Acute uraemia 10% vs. 9% N Not significant TIULA and
Chronic ureaemia 10% vs. 8.9% N Not indicated NEUVONEN
1986
Salicylate Alcoholic hepatitis 27% vs. 34% N Not known. BRODIE
Alcoholic cirrhosis 27% vs. 41% Y Bilirubin and and BooBIs
hypoalbuminaemia 1978
implicated in binding
defect
Drug Interactions at Plasma and Tissue Binding Sites 143
Table 1. Continued
Orug Disease % Free or free Significant Clinical significancel References
fraction (control difference comment
vs. patient)
Sulphadiazine Alcoholic hepatitis 46% vs. 58% Y Not known. Bilirubin BRODIE
Alcoholic cirrhosis 46% vs. 51 % N and hypoalbuminaemia and BoosTs
implicated in binding 1978
defect
Sulphisoxazole Uraemia 5.2% vs. 21.8% Not indicated. CALVO
Carbamylation of drug et a!. 1982
binding site I implicating in
defect; site II not affected
Theophylline Acute illness 54.6% vs. 69.7% Y Not significant as free ZAROWITZ
inCOPO 7.4% vs. 8.1 N concentration is not et a!. 1985
(I'glml) is not changed significantly
Tolfenamic acid Renal disease 0.08% vs. 0.17% Y Not clear. High affinity LAZNICEK
Liver disease 0.08% vs. 0.29% Y for red blood cells means and SENIUS
that these may act as 1986
reserve binding sites when
free fraction increases
Valproic acid Renal disease 8.4% vs. 20.3% Y Not clear. May lead to GUGLER
increased incidence of and
toxicity. Important in drug MUELLER
monitoring interpretation 1978
Early pregnancy' 9.4% vs. 11.5% Y Important in drug PERUCCA
Mid pregnancy 9.4% vs. 12.1 % Y monitoring. et a!. 1981
Late pregnancy 9.4% vs. 14.6% Y Oecreased albumin serum
concentration implicated in
binding defect
lOOM 6.2% vs. 7.6% Y Not significant if diabetes GAITI
is well controlled. If poorly et a!. 1987
controlled increased free
concentration may be
greater
Verapamil Arrhythmia Not indicated. MCGOWAN
Pharmacokinetic change et a!. 1982
may be implicated
Liver disease 0.099 vs. 0.16 Y Clinical end point effective GIACOMINI
to titrate dose so change et a!. 1984
not significant in therapy.
Change may be relevant
when interpreting kinetic
data and total drug
concentrations
Warfarin Chronic cardiac 1% vs.l% N No change expected FITCHL
failure et a!. 1983
Warfarin NIDOM 1.1 % VS. 1% N Not indicated (but no O'BYRNE
change expected) et a!. 1988
Zomepirac Uraemia 1.4% VS. 4% Y Decrease due to PRITCHARD
endogenous inhibitors et a!. 1983
Not clinically significant
Y, yes; N, no; MI, myocardial infarction; IHO, ischaemic heart disease; COPO, chronic obstructive pulmonary disease;
CV A, cerebrovascular accident; AGP, al-acid glycoprotein; K" drug-protein association constant; V d , apparent volume
of distribution; NIOOM, non-insulin-dependent diabetes mellitus; 100M, insulin-dependent diabetes mellitus.
, Although pregnancy is not a disease, it can give rise to changed binding; therefore it has been included in this table.
......
t
Is drug of interest> 90%
protein bound?
Yes
No
Yes
-------------=: ,
What is the hepatic Low Would a transient increase No
in free drug concentration 1-1- - - - - + Clinically significant interaction
extraction ratio not likely
of the drug? be clinically relevant?
High
Is the drug
given i.v.?
Yes
Yes
....
Clinically significant (1
interaction likely.
Perform a clinical study ~
C"l
to quantify effects Fig. 7. Algorithm for determining clinical significance of potential plasma tr1
protein binding displacement interactions. (After ROLAN 1994)
~
Drug Interactions at Plasma and Tissue Binding Sites 145
I. Conclusions
Plasma protein binding displacement has been overestimated and overstated
as a mechanism of drug-drug interactions since the effects are of a transient
nature; it is only considered to be problematic with regard to adverse clinical
outcomes under specific conditions. Drug interactions, which in the past have
been attributed to plasma binding displacement, have another underlying
mechanism, usually involving decreased elimination of the drug in the liver or
kidney. ROLAN (1994) has drawn up an algorithm to help assist in determining
whether displacement interactions are likely to be clinically significant. A copy
of this algorithm is presented in Fig. 7. If a plasma protein binding interaction
is suspected, and the drug has a narrow therapeutic range, it is advisable to
monitor free (unbound) concentrations of the drug as a guide to dosage
adjustment. This is also the case in patients who have diseases which give rise
to changed binding. Tissue binding displacement interactions, since re-equili-
bration of free drug concentrations post an interaction is dependent on the
drug's half-life, rather than on redistribution, can be more problematic and
this type of interaction can be complicated by decreased elimination of the
displaced drug, as is seen in the case of the classical interaction between
digoxin and quinidine.
References
Aggeler PM, O'Reilly RA, Leong L, Kowitz PE (1967) Potentiation of anticoagulant
effect of warfarin by phenylbutazone. N Engl 1 Med 276:496-501
Aguirre C, Rodriguez-Sasiain 1M, Calvo R (1994) Decrease in penbutolol protein
binding as a consequence of treatment with some alkylating agents. Cancer
Chemother Pharmacol 34:86-88
Allen LM, Creaven PI (1974) Binding of a new antitumour agent, thalicorpine to
DNA. 1 Pharm Sci 63:74-475
Barre J, Houin G, Brunner RF, Bree F, Tillement IP (1983) Disease induced modifica-
tions of drug pharmacokinetics. Int 1 Clin Pharmacol Res 3:215-226
Baruzzi A, Contin M, Perucca E, Albani F, Riva R (1986) Altered serum protein
binding of carbamazepine in disease states associated with an increased lX:t-acid
glycoprotein concentration. Eur 1 Clin Pharmacol 31:85-89
Beck WS, Dietzel K, Geisslinger G, Engler H, Vergin H, Brune K (1990) Effects of
sodium salicylate on elimination kinetics of indomethacin and bile production in
dogs. Drug Metab Dispos 18(6):962-967
Belpaire FM, Wynant P, Van Trappen P, Dhont M, Verstraete A, Bogaert MG (1995)
Protein binding of propranolol and verapamil enantiomers in maternal and foetal
serum. Br 1 Clin PharmacoI39:190-193
Bergrem H, Ritland S, Opendal I, Bergran A (1983) Prednisolone pharmacokinetics
and protein binding in patients with portosystemic shunt. Scand 1 Gastroenterol
18:273-276
Bigger IT (1979) The quinidine-digoxin interaction. What do we know about it? N Engl
1 Med 301:779-781
Bree F, Urien S, Nguyen P, Riant P, Albengres E, Tillement JP (1990) Are-evaluation
of the HSA-piroxicam interaction. Eur 1 Drug Metab PharmacoI15(4):303-307
Brodie LMl, Boobis D (1978) The effect of chronic alcoholic ingestion and alcoholic
liver disease on binding of drugs to serum proteins. Eur 1 Clin Pharmacol13:435-
438
146 J.C. McELNAY
Stewart CF, Fleming RA, Germain BF, Seleznick MJ, Evans WE (1991) Aspirin alters
methotrexate disposition in rheumatoid arthritis patients. Arthritis Rheum
34:1514-1520
Tillement JP (1980) Plasma binding of drugs. Pharm Int 1:64-65
Tillement JP, Lhoste F, Gidicelli JF (1978) Disease and drug protein binding. Clin
Pharmacokinet 3:144-154
Tiula E, Elfving S (1987) Serum protein binding of phenytoin, diazepam and
propranolol in age-related decrease in renal function. Ann Clin Res 129:163-169
Tiula E, Neuvonen PJ (1986) Effect of total drug concentration on the free fraction in
uraemic sera. Ther Drug Monit 8:27-31
Toon S, Low L, Gibaldi M, Trager WF, O'Reilly RA, Motley ChH (1986) The war-
farin-sulphinpyrazone interaction: stereochemical considerations. Clin Pharmacol
Ther 39:15-24
Trnavska Z, Krejocova H, Tkaczykovam, Salcmanova Z, Elis J (1991) Pharmacokinet-
ics of lamotrigine (Lamictal) in plasma and saliva. Eur J Drug Metab Pharmacokin
3:211-215
Webb D, Buss DC, Fifield R, Bateman DN, Routledge PA (1986) The plasma protein
binding of metoclopramide in health and renal disease. Br J Clin Pharmacol
21:334-336
Wood M, Shand DG, Wood AJ (1979a) Altered drug binding due to use of indwelling
heparinized cannulas (heparin lock) for sampling. Clin Pharmacol Ther 25:103-
107
Wood M, Shand DG, Wood AJ (1979b) Propranolol binding in plasma during cardio-
pulmonary bypass. Anesthesiol. 51:512-516
Zarowitz B, Shlom J, Eichenhorn MS, Popovich J (1985) Alternations in theophylline
protein binding in acutely ill patients and COPD. Chest 87:766-769
CHAPTER 5
Drug Interactions
and Drug-Metabolising Enzymes
P.F. D'ARCY
A. General Introduction
Induction and inhibition of cytochrome P450 enzymes are mechanisms that
underly some of the more serious drug-drug interactions; it is therefore perti-
nent that the structure, distribution and essential roles of cytochromes P450
are reviewed.
Cytochrome P450 is not a single species of protein; the system is actually
a collection of isoenzymes, all of which possess an iron atom in a porphyrin
complex. They catalyse different types of oxidation reactions and under cer-
tain circumstances may catalyse other types of reaction such as reduction
(TIMBRELL 1993).
Since the early experiments of CONNEY et al. (1956; CONNEY 1967) it has
often been observed that the rate of oxidative metabolism of various sub-
strates can differ markedly depending on the age, sex, species or the extent of
exposure of the animal to different inducing agents. A number of different
investigators have directed studies to evaluate the number and types of cyto-
chrome P450s that exist in a single organ. In particular they questioned
whether specific types of reactions catalysed by the microsomal electron trans-
port system required specific cytochrome P450, or whether many cytochrome
P450s have a rather broad substrate specificity differing only in the rate of
catalysis of each reaction.
SCHENKMAN (1993) has well documented the beginning of this interest and
study into the cytochrome P450 monooxygenase. It commenced with the
compilation of knowledge of the metabolism of xenobiotics by WILLIAMS
(1959), who described the many different metabolites produced from
xenobiotics in vivo by animals. From the late 1940s into the 1960s knowledge
was advanced by BRODIE et al. (1955), who was one of the first to start in vitro
biochemical studies on the metabolism of xenobiotics by oxidative enzymes.
Work from MILLER'S laboratory augmented these studies (MULLER and
MILLER 1953) and it soon became clear that liver micro somes were the source
of NADPH-dependent, oxidative enzymes capable of metabolising a number
of xenobiotics.
Since these early studies in the 1950s more than 800 different xenobiotics,
many of which are therapeutic drugs, have been shown to be substrates
for liver microsomal oxidative enzymes. The major types of oxidation
152 P.F. D'ARCY
aOther cytochrome P450s have been detected in animals; multiple gene conversions
have occurred among some animal CYP genes, rendering them quite dissimilar to their
counterparts in humans.
154 P.F. D'ARCY
c. Genetic Polymorphism
There are wide variations in cytochrome P450 activity in both humans and
animals. DALY and IDLE (1993) have suggested, in their comprehensive review
on animal and human cytochrome P450 polymorphisms, that this variation
may be due to either a genetic polymorphism which results in either lack of
protein synthesis or synthesis of a protein with reduced enzyme activity, or to
a variation in the level of cytochrom P450 gene expression due to regulation by
factors such as hormones or xenobiotics.
The inability of up to 12% of individuals from a variety of racial
groups to metabolise the antihypertensive agent debrisoquine to 4-
hydroxy-debrisoquine has been known since the 1970s (MAHGOUB et al. 1977).
Pedigree studies showed that the poor metaboliser phenotype was inherited
in an autosomal recessive manner (EVANS et al. 1980). Similar findings
have been reported for the antiarrhythmic and oxytocic agent sparteine
(EICHELBAUM et al. 1979), and other compounds including phenformin (OATES
et al. 1982) and bufuralol (DAYER et al. 1982). Further studies indicated that
a single enzyme was responsible for the metabolism of all these drugs
(BERTILSSON et al. 1980). At least 25 other compounds including beta-blockers,
antihypertensives, antiarrhythmics and antidepressants are known to be
metabolised by debrisoquine hydroxylase (see review by BROSEN and GRAM
1989).
Cloning and sequencing of the CYP2D genes has assisted identification of
the mutations which give rise to the debrisoquine polymorphism in humans,
and a number of variant alleles associated with the poor phenotype have been
identified. Debrisoquine phenotype may have important consequences with
regard to both response to drug therapy and exposure to xenobiotics. For
example, the use of the tricyclic antidepressant nortriptyline, the neuroleptic
perphenzazine, or the antiarrhythmics propafenone and flecainaide may cause
problems in both rapid extensive metabolisers (low and inadequate serum
concentrations) and in poor metabolisers (toxic serum concentrations)
(BROSEN and GRAM 1989). Fortunately, however, the majority of patients
exhibit a normal metabolism of these agents and gain maximum therapeutic
benefit with minimal toxicity.
Some interesting studies on a possible association between susceptibility
to lung cancer and debrisoquine metabolic phenotype have been carried out
by AYESH et al. (1984). They found that only 1.6% of a group of bronchial
carcinoma patients were poor metabolisers of debrisoquine compared
with 9.0% of a group of smoking controls. The molecular basis of this
possible association between the poor metaboliser phenotype and decreased
lung cancer susceptibility has been suggested by CRESPI et al. (1991) to relate
to a reduced ability to activate tobacco smoke-derived procarcinogens.
An association between the poor metaboliser phenotype and susceptibil-
ity to early-onset Parkinson's disease has been suggested by BARBEAU et al.
Drug Interactions and Drug-Metabolising Enzymes 157
I. Enzyme Inducers
Enzyme induction has been defined by GELEHRTER (1979) as an adaptive
increase in the number of molecules of a specific enzyme, secondary either to
an increase in its rate of synthesis or to a decrease in its rate of degradation. It
is believed that high substrate concentration, and not hormonal regulation, is
responsible for increased enzyme synthesis. If the demands on an enzyme
system exceed its maximal functional capacity, adaptation should entail a
parallel increase in enzymes and their containing structure.
Cytochrome P450 and its associated electron transport carriers are mem-
brane-bound proteins in mammalian tissue; thus the induction of hepatic
microsomal enzymes by phenobarbitone and other drugs has been shown
conclusively to stimulate an increase in the smooth endoplastic reticulum,
synthesis of enzyme protein, phospholipid and haem, DNA-dependent RNA
synthesis, and the synthesis of specific mRNAs (REMMER and MERKER 1963;
PARKE 1975; LINDELL et al. 1977). A further study of the effect of
phenobarbitone on rat liver also showed that there were no significant alter-
ations of the activities of any of the DNA-dependent RNA polymerases,
which suggested that the primary stimulus of enzyme induction was more
likely to be on post-transcriptional events such as enhanced stabilisation or
decreased turnover of hepatic RNA, than on the transcription of new RNA
(LINDELL et al. 1977).
Increased protein synthesis also appears to occur in the induction of the
hepatic microsomal mixed-function oxidases following treatment of animals
Drug Interactions and Drug-Metabolising Enzymes 159
then become the prime cause of their own inadequate levels in the blood. This
problem has particular relevance in, for example, the long-term treatment of
tuberculosis. ACOCELLA et al. (1972) have shown that rifampicin administered
constantly at 600mg/day for 7 days gave significantly lower blood levels on the
7th day than it did on the 1st day of treatment. Development of tolerance to
drug regimens is certainly caused, at least in part, by enzyme induction.
Because so many therapeutic drugs have been shown to be implicated in
cytochrome P450 induction, it is obviously impossible to describe or discuss
the effects of their interactions even concisely in a chapter of this type. As a
compromise therefore some examples of common P450-inducing agents are
given in Table 2, while some recent reports of serious drug-drug interactions
due to enzyme induction are shown as examples in Table 3.
In surveying the literature it soon becomes evident that reports of inter-
actions due to enzyme inhibition are far more numerous than those caused by
enzyme induction. This is not really surprising since the former are more
readily observed in the clinic and they evoke therapeutic overdosage or toxic-
ity, while the latter cause a reduced therapeutic effect of medication which
may not immediately be noticed or reported.
Alcohol
Chlorinated hydrocarbon pesticides
Polycyclic hydrocarbons (e.g., dioxin) and other environmental pollutants
Tobacco smoking
Many therapeutic drugs, including:
Barbiturates
Carbamazepine
Carbenoxolone
Ciprofioxacin
Clofibrate
Glucagon
Glucocorticoids (e.g., dexamethasone)
Isoniazid
Phenybutazone
Phenytoin
Primidone
Rifampicin
Theophylline
Drug Interactions and Drug-Metabolising Enzymes 161
(PAS), pheniprazine, the two narcotics pethidine and morphine, and the anti-
bacterial 4-quinolones. Some other agents, for example, chlorcyclizine,
glutethimide and phenylbutazone, can inhibit or induce drug metabolism
by enzymes depending upon whether they are administered acutely or chroni-
cally. Inhibition generally requires only a single dose of a compound rather
than repeated doses as does induction; the environmental impact of inhibition
is probably less than that of induction although inhibition may also be
relevant in the work place, for example, workers exposed to the solvent
dimethylformamide are less likely to suffer alcohol-induced flushes than those
not exposed, possibly due to the inhibition of alcohol metabolism (TIMBRELL
1993).
162 P.F. D'ARCY
Cobalt chloride
Industrial solvents (e.g., carbon tetrachloride, dimethylformamide)
Organophosphorus compounds
Many therapeutic drugs including:
Allopurinol
Amiodarone
Chloramphenicol
Cimetidine
Corticosteroids
Cyclophosphamide
Danazol
Dextropropoxyphene
Dicoumarol
Erythromycin
Felbamate
Fluconazole
Glibenclamide
Infiuenza vaccine
Iproniazid
Itraconazole
Ketoconazole
MAOI antidepressants
Moricizine
Nicoumalone
4-Quinolone and fiuoroquinolone antibacterials (e.g., ciprofioxacin, enoxacin,
lomefioxacin, norfioxacin, ofioxacin, perfioxacin)
Selegiline
Serotonin re-uptake inhibitor antidepressants (SRIs)
Tamioxifen
Triacetyloleandomycin
Drug Interactions and Drug-Metabolising Enzymes 163
Table 5. Serious drug-drug interactions due to enzyme inhibition (illustrative only, not
comprehensive)
F. Conclusions
It may be apparent from these brief accounts of interactions involving P450
enzyme induction or inhibition that many of the involved drugs are those on
which patients are carefully stabilised often for long periods of time (e.g.,
anticoagulants, antidepressants, hypoglycaemics, hypotensives, oral contra-
ceptives). Past experience has shown that it is these drug-stabilised patients
who are at special risk from any interaction that will influence the potency or
availability of their medication. This is especially so for the elderly patients,
whose medication needs are generally greater than younger persons and who
have a substantially greater risk than the younger patient of experiencing
adverse reactions to medication.
Drug Interactions and Drug-Metabolising Enzymes 165
References
Acocella G, Bonollo L, Garimoldi M, Mainardi M, Tenconi LT, Nicolis FB (1972)
Kinetics of rifampicin and isoniazid administered alone and in combination to
normal subjects and patients with liver disease. Gut 13:47-53
Adebayo GI (1988) Interaction between phenytoin and theophylline in healthy volun-
teers. Clin Exp Pharmacol Physiol 15:883-887
Ahmad SR (1992) USA antihistamine alert. Lancet 340:542
Alvares AP, Schilling G, Levin W, Kuntzman R (1967) Studies on the induction of CO-
binding pigments in liver microsomes by phenobarbital and 3-methylcholan-
threne. Biochem Biophys Res Commun 29:521-526
Angle M, Huff PS, Lea JW (1991) Interactions between oral contraceptives and thera-
peutic drugs. Outlook 9:1-6
Arin,< E (1993) Extrahepatic microsomal forms: lung microsomal cytochrome P450
isozymes. In: Schenkman JB, Greim H (eds) Cytochrome P450. Springer, Berlin
Heidelberg New York, pp 373-386
Armstrong G, Beg MF, Scahill S (1991) Warfarin potentiated by proguanil. Br Med J
3034:789
Axelrod J (1955) The enzymatic demethylation of ephedrine. J Pharmacol Exp Ther
114:430-438
Ayesh R, Idle JR, Ritchie JC, Crothers MJ, Hetzel MR (1984) Metabolic oxidation
phenotypes as markers for susceptibility to lung cancer. Nature 311:169-170
Barbeau A, Cloutier T, Roy M, Plasse L, Paris S, Poirier J (1985) Ecogenetics of
Parkinson's disease: 4-hydroxylation of debrisoquine. Lancet 11:1213-1216
Bertilsson L, Dengler HJ, Eichelbaum M, Schultz HU (1980) Pharmacogenetic
covariation of defective N-oxidation of sparteine and 4-hydroxylation of
debrisoquine. Eur J Clin PharmacoI17:153-155
Birnbaum LS (1993) Changes in cytochrome P450 in senescence. In: Schenkman JB,
Greim H (eds) Cytochrome P450. Springer, Berlin Heidelberg New York, pp 477-
492
Black SD (1993) Cytochrome P450 structure and function. In: Schenkam JB, Greim H
(eds) Cytochrome P450. Springer, Berlin Heidelberg New York, pp 156-168
Booth CD (1993) A drug interaction between danazol and warfarin. Pharm J 250:439-
440
Brodie BB, Axelrod J, Cooper JR, Gaudette LE, La Duc BN, Mitowa C, Udenfriend
S (1955) Detoxification of drugs and other foreign compounds by liver mi-
crosomes. Science 121:603-604
Brooks BA, McBride OW, Dolphin CT, Farrall M, Scambler PJ, Gonzalez FJ, Idle JR
(1988) The gene CYP3 encoding P45PCN1 (nifedipine oxidase) is tightly linked to
the gene COLIA2 encoding collagen type I alpha on 7q21-p22.1. Am J Hum Genet
43:280-284
Brosen K, Gram LF (1989) Clinical significance of the sparteine/debrisoquine oxida-
tion polymorphism. Eur J Clin PharmacoI36:537-547
Cadle RM, Zenon GJ III, Rodriguez-Barradas MC, Hamill RJ (1994) Fluconazole
induced symptomatic phenytoin toxicity. Ann Pharmacother 28:191-195
Cashman JR, Hanks D, Weiner RI (1987) Epoxy derivatives of arachidonic acid are
potent stimulators of prolactin secretion. Neuroendocrinology 46:245-251
Castel JM, Cappiello E, Leopaldi D, Latini R (1990) Rifampicin lowers plasma
concentrations of propafenone and its antiarrhythmic effect. Br J Clin Pharmacol
30:155
Committee on Safety of Medicines (1989) Serious interaction between tamoxifen and
warfarin. Committee on Safety of Medicines, London (Current problems, no 26)
Conney AH (1967) Pharmacological implications of microsomal enzyme induction.
Pharmacol Rev 19:317-366
Conney AH, Miller EC, Miller JA (1956) The metabolism of methylated aminoazo
dyes. V. Evidence for induction of enzyme synthesis in the rat by 3-methylcholan-
threne. Cancer Res 16:450-459
Drug Interactions and Drug-Metabolising Enzymes 167
Conney AH, Miller EC, Miller JA (1957) Substrate-induced synthesis and other
properties of benzpyrenehydroxylase in rat liver. J BioI Chern 228:753-766
Consumers' Association (1993) Fluoroquinolones reviewed. Drug Ther Bull
31(18):69-72
Crespi CL, Penman BW, Gelboin HV, Gonzalez FJ (1991) A tobacco smoke-derived
nitrosamine 4-(methylnitrosamino)-I-(3-pyridyl)-I-butanone is activated by
multiple human cytochrome P450s including the polymorphic human cytochrome
P4502D6. Carcinogenesis 12:1197-1201
Daly AK, Idle JR (1993) Genetics: animal and human cytochrome P450 polymor-
phisms. In: Schenkman JB, Grein H (eds) Cytochrome P450. Springer, Berlin
Heidelberg New York, pp 433-446
Damani LA (1988) The flavin-containing monooxygenase as an amine oxidase. In:
Gorrod JW, Oelschlager H, Caldwell J (eds) Metabolism of xenobiotics. Taylor
and Francis, London, pp 59-70
D'Arcy PF (1984a) Tobacco smoking and drugs: a clinically important interaction?
Drug Intell Clin Pharm 18:302-307
D'Arcy PF (1984b) Vaccine-drug interactions. Drug Intell Clin Pharm 18:697-700
D'Arcy PF (1986) Drug interactions with oral contraceptives. Drug Intell Clin Pharm
20:353-362
D'Arcy PF, Merkus FWHM (1981) Alcohol and drug interactions. Pharm Int 2:273-
280
Davis GC, Williams AC, Markey SP, Ebert MH, Caine ED, Reichert CM, Kopin IJ
(1979) Chronic parkinsonism secondary to intravenous injection of meperidine
analogues. Psychiatry Res 1:249-254
Dayer P, Balant L, Courvoisier F, Kupfer A, Kubli A, Georgia A, Fabre J (1982) The
genetic control ofbufuralol metabolism in man. Eur J Drug Metab Pharmacokinet
7:73-77
Dean WL, Gray RD (1982) Relationship between state of aggregation and catalytic
activity for cytochrome P-450 LM2 and NADPH-cytochrome P-450 reductase.
J BioI Chern 257:14679-14685
DeWaziers I, Cugnenc P, Yang CS, Leroux JP, Beaune P (1990) Cytochrome P-450
isozymes, epoxide hydrolase and glutathione transferase in rat and human hepatic
and extrahepatic tissues. J Pharmacol Exp Ther 253:387-394
Dillard ML, Fink RM, Parkerson R (1992) Ciprofloxacin-phenytoin interaction. Ann
Pharmacother 26:263
Eichelbaum M, Spannbrucker N, Steincke B, Dengler HJ (1979) Defective N-oxidation
of sparteine in man: a new pharmacogenetic defect. Eur J Clin PharmacoI17:153-
155
Elofsson S-A, Gamberale F, Hindmarsh T, Iregren A, Isaksson A, Johnsson I, Knave
B, Lydahl E, Mindus P, Persson HE, Philipson B, Steby M, Struwe G, SOderman
E, Wennberg A, Widen L (1980) Exposure to organic solvents. A cross-sectional
epidemiological investigation on occupationally-exposed car and industry spray
painters with special reference to the nervous system. Scand J Work Environ
Health 6:239-273
Estabrook RW (1979) Concluding remarks. In: Estabrook RW, Lindenlaub E (eds)
The induction of drug metabolism. Schattauer, Stuttgart, pp 643-645 (Symposia
Medica Hoechst, vol 14)
Evans DAP, Mahgoub A, Sloan TP, Idle JR, Smith RL (1980) A family and population
study of the genetic polymorphism of debrisoquine oxidation in a British white
population. J Med Genet 17:102-105
Gannon RH, Anderson ML (1992) Fluconazole-nortriptyline drug interaction. Ann
Pharmacother 26:1456-1457
Gelehrter TD (1979) Enzyme induction in mammals - an overview. In: Estabrook RW,
Lindenlaub E (eds) The induction of drug metabolism. Schattauer, Stuttgart, pp 7-
24
Gonzalez FJ (1993) Cytochrome P450 in humans. In: Schenkman JB, Greim H (eds)
Cytochrome P450. Springer, Berlin Heidelberg New York, pp 239-257
168 P.F. D'ARCY
Monig H, Baese C, Heidemann HT, Ohnhaus EE, Schulte HM (1990) Effect of oral
contraceptive steroids on the pharmacokinetics of phenprocoumon. Br J Clin
PharmacoI30:115-116
Mi.iller GC, Miller JA (1953) The metabolism of methylated aminoazo dyes. II.
Oxidative demethylation by rat liver homogenates. J BioI Chern 202:579-
587
Murphy RC, Falk JR, Lumni S, Yadagari P, Zirrolli JA, Balazy M, Masferrer JL,
Abraham NG, Schwartzman ML (1988) 12(R)-Hydroxyeicosatrienoic acid: a
vasodilator cytochrome P450-dependent arachidonate metabolite from bovine
corneal epithelium. J BioI Chern 263:17197-17202
Nebert DW, Gonzalez EJ (1985) Cytochrome P-450 gene expression and regulation.
Trends Pharmacol Sci 6:160-164
Noticeboard (1992) Warning about astemizole. Lancet 340:1155
Oates S, Shah RR, Idle JR, Smith RL (1982) Genetic polymorphism of phenformin 4-
hydroxylation. Clin Pharmacol Ther 32:81-89
Pantuck EJ, Hsiao KC, Kuntzman R, Conney AH (1975) Intestinal metabolism of
phenacetin in the rat: effect of charcoal-broiled beef and rat chow. Science
187:744-745
Parke DV (1975) Induction of the drug-metabolising enzymes. In: Parke DV (ed)
Enzyme-induction. Plenum, London, p 207
Parke DV (1979) The responsiveness of cells to various drug inducers. In: Estabrook
RW, Lindenlaub E (eds) The induction of drug metabolism. Schattauer, Stuttgart,
pp 105-111
Philpot RM, Wolf CR (1981) The properties and distribution of the enzymes of pulmo-
nary cytochrome P450 dependent monooxygenase systems. In: Hodgson E, Bend
JR, Philpot RM (eds) Reviews in biochemical toxicology, vol 3. Elsevier,
Amsterdam, pp 51-76
Philpot RM, Arin,< E, Fouts JR (1975) Reconstitution of the rabbit pulmonary microso-
mal mixed-function oxidase system from solubilized components. Drug Metab
Dispos 3:118-126
Posner HS, Mitoma C, Udenfriend S (1961) Enzymatic hydroxylation of aromatic
compounds. II. Further studies of the properties of the microsomal hydroxylating
system. Arch Biochem Biophys 94:269-279
Remmer H, Merker HJ (1963) Drug induced changes in the liver endoplasmic
reticulum: association with drug metabolizing enzymes. Science 14:1657-
1658
Rennert H, Fischer RT, Alkvarez JG, Trzaskos JM, Strauss JM (1990) Generation of
regulatory oxysterols: 26-hydroxylation of cholesterol by ovarian mitochondria.
Endocrinology 127:738-746
Sacerdoti D, Abraham NG, McGiff JC, Schwartzman ML (1988) Renal cytochrome
P450-dependent metabolism of arachidonic acid in spontaneously hypertensive
rats. Biochem PharmacoI37:521-527
Schenkman JB (1993) Historical background and description of the cytochrome P450
monooxygenase system. In: Schenkman JB, Greim H (eds) Cytochrome P450.
Springer, Berlin Heidelberg New York, pp 3-13
Schmucker DL, Woodhouse KW, Wang RK, Wynne H, James OF, McManus M,
Kremers P (1990) Effects of age and gender on in vitro properties of human liver
microsomal monooxygenases. Clin Pharmacol Ther 48:365-374
Seaton TL, Ceklum CL, Back DJ (1991) Possible potentiation of warfarin by
fluconazole. Ann Pharmacother 24:1177-1178
Serpa MD, Cossolias J, McCreevy MJ (1992) Moricizine-warfarin: a possible drug
interaction. Ann Pharmacother 26:127
Shaeffer MS, Collier D, Sorrell MF (1994) Interaction between FK 506 and erythromy-
cin. Ann Pharmacother 28:280-281
Shimada T, Guengerich FP (1989) Evidence for cytochrome P-450NF, the nifedipine
oxidase, being the principal enzyme involved in the bioactivation of aflatoxins in
human liver. Proc Natl Acad Sci USA 86:462-465
170 P.F. D'ARCY
Sladek NE, Mannering GJ (1966) Evidence for a new P-450 hemoprotein in hepatic
microsomes from methylcholanthrene treated rats. Biochem Biophys Res
Commun 24:668-674
Snyder GD, Capdevila J, Chacos N, Manna S, Flack JR (1983) Action of luteinizing
hormone-releasing hormone: involvement of novel arachidonic acid metabolites.
Proc Nat! Acad Sci USA 80:3504-3507
Soto J, Sacrisan JA, Arellano F, Mazas J (1991) Possible theophylline-amiodarone
interaction. Ann Pharmacother 24:1115
Srivastava SP, Seth PK (1983) 7-Ethoxycoumarin O-deethylase activity in rat brain
microsomes. Biochem Pharmacol 32:3657-3660
Staum JM (1990) Enzyme induction: rifampicin-disopyramide interaction. Ann
Pharmacother 24:701-703
Strobel HW, Stralka DJ, Hammond DK, White T (1993) Extrahepatic microsomal
forms: gastrointestinal cytochromes P450, assessment and evaluation. In:
Schenkman JB, Greim H (eds) Cytochrome P450. Springer, Berlin Heidelberg
New York, pp 363-371
Tenni P, Lalich DL, Byrne MJ (1989) Life threatening interaction between tamoxifen
and warfarin. Br Med J 298:93
Timbrell JA (1993) Biotransformation of xenobiotics. In: Ballantyne B, Marrs T,
Turner P (eds) General and applied toxicology, voll. Macmillan, Basingstoke, pp
89-119
Tisdel KA, Israel D SA, Kolb KW (1994) Warfarin-felbamate interaction: first report.
Ann Pharmacother 28:805
Tjia JF, Back DJ, Breckenridge AM (1989) Calcium channel antagonists and
cyclosporin metabolism: in vitro studies with human liver microsomes. Br J Clin
PharmacoI28:362-365
Volk B, Amelized Z, Anagnostopoulos J, Knoth R, Oesch F (1988) First evidence of
cytochrome P450 induction in the mouse brain by phenytoin. Neurosci Lett
84:219-224
Warner A, Gustafsson J-A (1993) Extrahepatic microsomal forms: brain cytochrome
P450. In: Schenkman JB, Greim H (eds) Cytochrome P450. Springer, Berlin
Heidelberg New York, pp 387-397 0
Zeigler DM (1985) Molecular basis for N-oxygenation of sec- and tert-amines. In:
Gorrod JW, Damani LA (eds) Biological oxidation of nitrogen in organic mol-
ecules. Ellis Horwood, Chichester, pp 45-52
Zornberg GL, Bodkin JA, Cohen BA (1991) Severe adverse interactions between
pethidine and selegiline. Lancet 337:246
CHAPTER 6
Drug Interactions
Involving Renal Excretory Mechanisms
A. SOMOGYI
A. Introduction
The kidney represents the final elimination organ for virtually all foreign
substances irrespective of whether they are cleared unchanged by the kidney
or whether as metabolites formed in the body (predominantly the liver). It had
long been regarded that filtration at the glomerulus was the sole excretory
mechanism in the kidney for foreign substances. However, in 1874,
Heidenhain demonstrated the phenomenon of secretion by canine tubular
cells using the dye indigo carmine, and Marshall and colleagues in the 1920s
conclusively showed secretion of organic anions by the proximal tubules. Thus
the concept of filtration and secretion by the kidney came into existence. In
1929 M~ller and co-workers coined the term clearance, in which they math-
ematically described the excretion of substances by the kidney. It is
somewhat surprising that the concepts developed by these early pioneers of
renal physiology are still highly applicable in pharmacokinetics today. The
active tubular secretion of organic cations was shown for the first time by
Rennick in the dog using tetraethylammonium and by Sperber using a variety
of organic cations in the chicken. Further details of the history of renal physi-
ology/pharmacology can be found in several reviews (PETERS 1960; WEINER
and MUDGE 1964; RENNICK 1972).
allows some materials to pass through whilst restricting the passage of others.
The resulting filtrate is free of protein and substances with a molecular weight
of greater than 40000 and about 20A in diameter. The rate of glomerular
filtration is about 1801/day or 120ml/min.
The pharmacokinetic consequence of glomerular filtration is that the
clearance rate of drugs at the glomerulus [CL(GFR)] can be approximated to
the unbound fraction (fu) in plasma of the drug multiplied by the glomerular
filtration rate (GFR).
CL(GFR) = fu x GFR
Hence for drugs which are highly bound (fu < 0.1) in plasma, clearance at the
glomerulus is substantially less than the glomerular filtration rate, and the
maximum clearance of any drug at the glomerulus will be equal to the glom-
erular filtration rate.
The mechanisms by which drugs can interact at the glomerulus are: (a)
displacement from protein-binding sites. The pharmacokinetic consequence
will be that fu will increase and clearance of the drug by the glomerulus will
also increase in direct proportion to the increase in fu. There are very few
examples in the literature of drug-drug interactions occurring exclusively at
the glomerulus. (b) Where the glomerulus is physically damaged through the
nephrotoxic action ofaxenobiotic. In this case, the maximum clearance of a
drug will be equal to the GFR, irrespective of the value of the drug's fu.
However, this type of interaction is poorly documented and would rarely
occur on its own.
1. Physiological Considerations
The blood flow to the proximal tubule via the renal arteries is approximately
85% of total renal blood flow. Thus the maximum clearance of any drug
by tubular secretion at this site is equivalent to the total renal blood flow of
lo21/min.
2. Cellular Mechanisms
Our increasing understanding of the cellular mechanisms of transport of drugs
across the epithelial membranes of the proximal tubule has been accomplished
Drug Interactions Involving Renal Excretory Mechanisms 175
I. Organic Anions
The most commonly investigated interacting drug has been probenecid with
many published studies showing that it reduces the renal clearance of drugs
which are organic anions.
1. Probenecid
It was the discovery of penicillin by Lord Florey which ultimately led to the
discovery of probenecid. Penicillin although effective had a short duration of
action due to its very high renal clearance and short half-life. A number of
strategies were investigated to overcome this therapeutic problem. One strat-
egy was to try and block the renal excretion of penicillin. The development of
probenecid (BEYER et al. 1951) heralded the clinical practice of elevating blood
concentrations of organic anion drugs by blocking their renal tubular secre-
tion. Since then, probenecid has been used as a prototypic in vitro and in vivo
probe to assess whether an organic anion drug is secreted by the proximal
tubules. This has been documented for a number of drugs but, in particular,
penicillin and cephalosporin antibiotics. In this review, the symbol ± always
refers to the standard deviation.
178 A. SOMOGYI
a) Allopurinol
EUON et al. (1968) showed in three patients with gout that the administration
of 1 g probenecid increased the renal clearance of oxypurinol (the active
metabolite of allopurinol) from about 12ml/min to 36ml/min, without any
change in inulin clearance (a measure of GFR). The mechanism for this
increase in renal clearance involves inhibition by probenecid of the active
luminal re-uptake of oxypurinol, via the uric acid transport system.
c) Frusemide
Frusemide is an organic acid, highly protein bound (unbound fraction is less
than 0.1), loop diuretic which produces its pharmacological effect(s) at the
luminal rather than the peritubular site of the renal tubule. Access to the
luminal site is mainly by active secretion by the organic anion transport sys-
tem. It was hypothesized that administration of probenecid could blunt the
diuretic and natriuretic effect of frusemide by blocking access to its site of
action. Four healthy normal subjects received a single intravenous injection of
1 mg/kg frusemide with and without pretreatment of O.S g probenecid 8 and
2 h before the frusemide dose. Probenecid reduced the renal clearance of
frusemide from 134 ± 23 to 63 ± 6mllmin (P < O.OS). However, there was no
significant difference in the 6-h urine volume or sodium excretion (HONARI et
al. 1977). When frusemide was administered as an intravenous infusion in four
subjects, the reduction in frusemide renal clearance was accompanied by a
rise in plasma frusemide concentrations, but urine flow rate (29 ± 4 ml/min
frusemide alone and 15 ± 4mllmin with probenecid) and excreted fraction of
filtered sodium (19% ± 7% alone and 14% ± 9% with probenecid) declined
significantly (P < 0.05) (HONARI et al. 1977). In six normal, healthy subjects, the
effect of probenecid (0.5 g every 6 h for 3 days) on the disposition and effect
of frusemide 40mg intravenously was evaluated (HOMEIDA et al. 1977).
Probenecid significantly (P < 0.01) reduced the renal clearance of frusemide
from 90 ± 24 to 20 ± 12ml/min. In the first 30min after frusemide administra-
tion, there was a significant (P < 0.01) reduction in sodium excretion rate from
3.1 ± 0.6 to 2.2 ± 0.7mmollmin. However, there was no difference in the ratio
between the urinary frusemide concentration and urinary sodium concentra-
Drug Interactions Involving Renal Excretory Mechanisms 179
d) Indomethacin
BABER et al. (1978), in a study in 17 patients with rheumatoid arthritis, admin-
istered 25 mg indomethacin three times a day and demonstrated that
probenecid 0.5 g twice a day significantly increased plasma concentrations of
indomethacin by an average of 63%, but there was no difference (P> 0.05) in
the renal clearance of indomethacin (3.0 ± 2.5 mllh per kilogram indomethacin
alone versus 2.7 ± 2.5mllh per kilogram indomethacin plus probenecid). How-
ever, probenecid significantly (P < 0.01) reduced the apparent renal clearance
of the glucuronide conjugate of indomethacin from 271 ± 158 mllmin to 126 ±
188ml/min. This study highlighted a new type of renal drug interaction, in
which the renal clearance of a conjugated metabolite rather than the parent
drug was inhibited by another drug via competition for active tubular secre-
tion. However, it is not known whether the glucuronidation of indomethacin
occurs in the kidney (proximal tubule cells) and whether probenecid blocks
access of indomethacin to the site of renal glucuronidation or inhibits renal
glucuronidation.
e) Clofibrate
In four healthy male subjects who received clofibrate (500mg orally every
12h) with and without 6 hourly probenecid, steady-state concentrations of
unbound clofibric acid were increased almost fourfold (VEENENDAAL et al.
1981). The renal clearance of unbound clofibric acid was significantly (P <
0.05) reduced by probenecid from 14 ± 5 to 3 ± 1 ml/min, which is consistent
with inhibition of tubular secretion.
f) Sulphinpyrazone
In four patients with gout, the administration of probenecid reduced the renal
clearance of sulphinpyrazone from 21 ± 11 to 8 ± 5 ml/min, and when the
180 A. SOMOGYI
degree of protein binding (fu <0.02) was considered, the renal tubular secre-
tion of this organic anion was inhibited by a mean of 75% (as inulin clearance
remained unchanged) (PEREL et al. 1969).
g) Zidovudine
This antiviral agent undergoes extensive glucuronidation to form the ether
glucuronide GAZT (3'-azido-3' -deoxy-5'-~D-glucopyranuronosylthymidine)
(DE MIRANDA et al. 1989). In seven patients with AIDS or AIDS-related
complex, the administration of probenecid 500mg every 4h substantially al-
tered the disposition of oral zidovudine (2mg/kg 8 hourly). Probenecid signifi-
cantly (P < 0.01) increased the area underneath the plasma concentration-time
profile of zidovudine from 0.97 ± 0.04 to 2.00 ± O.77mg/1 x h. In addition, the
area underneath the plasma concentration-time curve of the glucuronide me-
tabolite was also significantly increased by probenecid from 6.9 ± 2.0 to 14.0 ±
5.7mg/1 x h (P < 0.01). In four of the subjects from whom urine could be
collected, the renal clearance of zidovudine was not altered by probenecid
(188 ± 65 vs. 141 ± 31mllmin per 1.73m2). However, the apparent renal
clearance of the glucuronide metabolite was substantially and significantly
reduced (P < 0.01) by probenecid from 293 ± 46 to 114 ± 50mllmin per 1.73m2
(Fig. 2). In a pilot study in two healthy subjects (HEDAYA et al. 1990),
probenecid 500mg every 6h for 3 days reduced the renal clearance of
zidovudine from 4.38 and 5.13mllmin per kilogram to 2.68 and 3.27mllmin per
kilogram, respectively. For the glucuronide conjugate, probenecid substan-
tially reduced the renal clearance from 11.6 and 11.0 to 2.82 and 2.44mllmin
per kilogram, respectively. Both groups attributed their findings to inhibition
5
--0-- GAZT PRE·PROBENECID
--0- GAZT POST·PROBENECID
4
o~~~~~-=~~~~~
o 2 4 6 8 10
Time (hours)
Fig. 2. Plasma concentration-time profile of the glucuronide conjugate of zidovudine
(GAZT) following administration of zidovudine alone (pre-probenecid) and after
concomitant administration of probenecid (post-probenecid) in seven patients.
Probenecid significantly reduced the apparent renal clearance of GAZT. (From DE
MIRANDA et al. 1989)
Drug Interactions Involving Renal Excretory Mechanisms 181
h) Acyclovir
In three patients, LASKIN et al. (1982) showed that probenecid 1 g orally 1 h
before acyclovir (administered as an intravenous infusion of 5mg/kg) signifi-
cantly (P < 0.05) reduced the renal clearance of acyclovir from 248 ± 80 to 168
± 48mllmin per 1.73m2 , with no alteration in creatinine clearance (a measure
of GFR). Acyclovir is both a weak acid (pKa 2.3) and weak base (pKa 9.3) and
it is likely, given the inhibition shown with probenecid, that the tubular secre-
tion of acyclovir is predominantly via the organic anion transporter.
i) Penicillins
Since the discovery that probenecid could increase serum penicillin concentra-
tions in humans (BURNELL and KIRBY 1951), many studies (too numerous to
cite) have shown that the renal clearance and hence tubular secretion of those
penicillins which are secreted can be reduced by probenecid, resulting in
elevated plasma concentrations. In a comprehensive study into the renal tubu-
lar secretion of benzylpenicillin and its inhibition by probenecid (OVERBOSCH
et al. 1988), four healthy volunteers underwent three different studies. In each
study, benzylpenicillin was infused to achieve three different steady-state
plasma concentrations. In the second and third studies, probenecid was admin-
istered by continuous infusion at low (31.3 mg/h in one subj ect, 21.4 mg/h in the
other three subjects) and high (between 93.8 and 200.3mg/h) doses resulting
in plasma probenecid concentrations of 6.7 and 35.1 mg/l, respectively.
Probenecid, in a concentration-dependent manner, increased the unbound
plasma concentrations of benzylpenicillin with an estimated probenecid ECso
of 52.3mg/1. When benzylpenicillin was administered alone at the three differ-
ent infusion rates, the data were able to be fitted to a non-linear equation in
which the maximum tubular excretion rate for benzylpenicillin (range 2493-
4131 mg/h) and ECso (plasma benzylpenicillin concentration at which half the
maximum excretion rate was achieved) of 22.6 to 60.5 mg/l could be derived.
When probenecid was administered in studies 2 and 3, the EDso (probenecid
dose at which 50% of the tubular transport is inhibited) was estimated to be
between 13.2 and 108.5 mg/h. These investigators extrapolated their data to
the clinical setting and suggested that the commonly used daily dose of 2 g/day
of probenecid is likely to be close to the maximum effective dose for inhibition
of the tubular secretion of benzylpenicillin.
Probenecid has also been shown to reduce the renal clearance of nafcillin
from 2.0 ± 0.5 mllmin per kilogram to 0.56 ± 0.17 mllmin per kilogram (P <
182 A. SOMOGYI
0.05) (WALLER et al. 1982) in five healthy subjects, and increase ticarcillin
plasma concentrations (CORVAIA et al. 1992); penicillins which have a renal
clearance greater than clearance due to glomerular filtration alone.
Of all the drugs interacting with probenecid, inhibition of the renal tubular
secretion of penicillins is considered to be the prototypic interaction for drugs
involving the kidney.
j) Quinolone Antibiotics
Several of these antibiotics are predominantly cleared by the kidney with renal
clearance values in excess of the glomerular filtration rate, indicating net
tubular secretion. In eight healthy male subjects, probenecid significantly (P <
0.01) reduced the renal clearance of cinoxacin to creatinine clearance ratio
from 1.32 ± 0.48 to 0.32 ± 0.17 (RODRIGUEZ et al. 1979). Since cinoxacin is
only 40% unbound in plasma, these data indicate that there is little tubular
reabsorption. Similar results were reported by ISRAEL et al. (1978), in which
probenecid reduced the renal clearance of cinoxacin from 153 ± 60mllmin per
1.73m2 to 66 ± 9mllmin per 1.73m2 (P < 0.001) in four healthy subjects. For
ciprofloxacin, with a renal clearance of over 200 ml/min, probenecid has been
shown to reduce its renal clearance by approximately 50% in healthy subjects
(WINGENDER et al. 1985); however, for fleroxacin, which has a renal clearance
of unbound drug of about 140ml/min, probenecid had no effect on its renal
clearance in five healthy subjects (WEIDEKAMM et al. 1987). These results again
support the hypothesis that for this class of antibiotic, if renal clearance is
greater than clearance by filtration, then tubular secretion is by, at least in part,
the organic anion transport system.
k) Cephalosporin Antibiotics
Interactions between cephalosporin antibiotics and probenecid have been
comprehensively reviewed by BROWN (1993). A large number of studies have
been published investigating the effect of probenecid on the disposition of the
cephalosporins. Some studies have shown a significant interaction to occur in
which plasma concentrations are increased, whilst others have not. The two
criteria for an interaction to occur in the kidney are, firstly, the kidney must
eliminate a large proportion of the drug. For the cephalosporins this can range
from 40% for ceftriaxone to 95% for cefuroxime (BROWN 1993). The second
criterion is that the renal clearance of the drug must exceed the clearance
attributed to filtration at the glomerulus. For some cephalosporin antibiotics
such as moxalactam, renal clearance does not exceed the glomerular filtration
clearance and hence probenecid has no effect on their renal clearance (Table
1). Two studies will be cited to illustrate the effects of probenecid on the renal
disposition of cephalosporins. In eight healthy male subjects, the effect of
probenecid 500 mg twice daily for 2 days prior to and on the morning of the
study was investigated to determine its effect on the disposition of cephradine
given as a 500-mg intravenous dose (ROBERTS et al. 1981). For cephradine,
Drug Interactions Involving Renal Excretory Mechanisms 183
90% of the dose is excreted unchanged in urine, and in normal healthy subjects
renal clearance is approximately 400ml/min and the degree of plasma binding
is low. Hence this drug's major clearance is by tubular secretion. Probenecid
significantly (P < 0.01) increased the area under the serum cephradine concen-
tration-time curve from 24 ± 9 to 57 ± 11 mg/lx h. This was entirely due to a
significant (P < 0.01) reduction in renal clearance from a mean of 221/h to
12l/h. Interpretation of the data is consistent with inhibition of the renal
tubular secretion of cephradine by probenecid via the organic anion transport
system. As a second example, the disposition of cefonicid (500mg intramuscu-
larly) was investigated when probenecid (1 g orally) was administered to eight
healthy subjects (PITKIN et al. 1981). Approximately 90% of the dose of
cefonicid is excreted unchanged in urine, and its renal clearance is only ap-
proximately 30mllmin. However, this antibiotic is highly bound in plasma,
with an unbound fraction of 0.05. Hence although the renal clearance is low it
is mainly via tubular secretion. Probenecid significantly (P < 0.001) increased
the area under the cefonicid plasma concentration-time curve from 346 ±
34mg/1 x h to 724 ± 99mg/1 x h. The terminal half-life of cefonicid was
prolonged from 4.9 ± 0.8h to 7.5 ± 1.4h and the renal clearance of cefonicid
184 A. SOMOGYI
was substantially reduced in the 4- to 6-h time period from 30.2 ± 7.1 mllmin to
7.8 ± 2.0ml/min (P < 0.001) (PITKIN et al. 1981). The investigators calculated
that the secretion rate of cefonicid was reduced by probenecid from a mean of
744 to 246.ug/min. These data are again entirely consistent with inhibition
of renal tubular secretion. Table 1 is a summary of data on the effect of
probenecid on the renal disposition of the cephalosporin antibiotics. In all
cases, cephalosporin antibiotics whose disposition (hence renal clearance)
is altered by probenecid have renal clearance values indicating net tubular
secretion.
/) Famotidine
This organic cation histamine Hz antagonist has a renal clearance in humans of
almost three times the glomerular filtration rate, indicating net renal tubular
secretion presumably by the organic cation transport system. In eight young
healthy male volunteers (INOTSUME et al. 1990) the administration of
probenecid 1000mg 2h before, 250mg 1 h before and 250mg with a 20-mg oral
dose of famotidine resulted in the area underneath the plasma famotidine
concentration-time curve being significantly increased (P < 0.01) from 424 ±
54ng/ml x h to 768 ± HOng/ml x h. The renal clearance of famotidine was
reduced from 297 ± 54mllmin to 107 ± 14ml/min (P < 0.01). Using the
famotidine unbound fraction in plasma and creatinine clearance values, these
authors calculated that the clearance of famotidine due to tubular secretion
was significantly (P < 0.01) reduced by 89% (from 196 ± 61mllmin to 22 ±
12mllmin). The mechanism of this interaction is unclear as famotidine is an
organic cation, whereas probenecid's inhibitory effect is considered to be
specific for organic anions. It may well be that probenecid is a polysubstrate
which may interact with the organic anion transport system and suitable
substrates of the organic cation transport system. This has been investigated in
vitro with similar substances (ULLRICH et al. 1994).
2. Methotrexate
The antineoplastic and antirheumatoid drug methotrexate is approximately
50% excreted unchanged in urine (depending on the dose). Its renal clearance
is approximately 75 mllmin and the unbound fraction in plasma is 0.5. It has
been shown that methotrexate, being an organic anion undergoes renal tubu-
lar secretion by the organic anion transport system (LIEGLER et al. 1969) and
there is also some evidence for active tubular reabsorption (HENDEL and
NYFORS 1984). Although its renal clearance increases with urine pH, suggestive
of passive reabsorption, this was attributed to greater solubility of the drug in
urine (SAND and JACOBSEN 1981). Several drug interactions have been re-
ported involving the renal clearance of methotrexate. Since the drug has a
narrow therapeutic index, these interactions have sometimes been manifested
in overt methotrexate toxicity. Drugs which interact renally with methotrexate
are reviewed below.
Drug Interactions Involving Renal Excretory Mechanisms 185
a) Probenecid
In four patients administered intravenous methotrexate 200mg/m2 , the co-
administration of probenecid (500-1000mg) resulted in serum concentrations
at 24h which were four times higher (mean = OAmg/l) than with four other
patients not receiving probenecid (mean = 0.09mg/l) (AHERNE et a1. 1978). The
renal clearance of methotrexate was 104 ± 34ml/min, which was significantly
(P < 0.05) reduced to 46 ± 15 mllmin in those patients also taking probenecid.
In a more comprehensive study, three patients receiving high-dose metho-
trexate (3.0 g/m2) also received probenecid (dose not stated, but serum con-
centrations were between 21 and 116mg/l) (HOWELL et a1. 1979). There was a
significant increase in serum methotrexate concentrations, but, as only three
subjects were studied, the overall disposition of methotrexate was
not statistically significantly altered. Nevertheless, at 24h after the dose
of methotrexate, probenecid resulted in a threefold increase in plasma
methotrexate concentrations. In addition, in CSF at both 24 and 72h,
methotrexate concentrations were increased by a factor of 2.8- and 4.2-fold,
respectively. The half-life of methotrexate in CSF was not altered by
probenecid. This is a clinically important interaction as methotrexate toxicity
(bone marrow depression) has been reported in some patients receiving
probenecid. In the monkey, probenecid has been shown to completely inhibit
the renal tubular transport of methotrexate, resulting in renal clearance values
being equivalent to filtration at the glomerulus (BOURKE et a1. 1975).
b) Penicillins
There have been no detailed pharmacokinetic studies investigating the inter-
action between methotrexate and penicillins. However, a number of case
reports involving different penicillins have shown marked (penicillin by 35%,
piperacillin by 66%, ticarcillin by 40%, dicloxacillin by 93%) reductions in the
clearance of methotrexate (BLOOM et a1. 1986). The clinical importance of this
interaction was highlighted in patients receiving low-dose methotrexate for
psoriasis (MAYALL et a1. 1991). Five patients taking between 7.5 and 12.5mg
methotrexate daily developed neutropenia and thrombocytopenia when also
receiving amoxycillin, flucloxacillin, benzylpenicillin or piperacillin. Three of
these patients died. The clinical seriousness of this interaction highlights the
importance of drug interactions involving renal tubular secretion mechanisms.
c) Urinary Alkalinizers
Methotrexate can also undergo passive renal tubular reabsorption depending
on urine pH and the degree of ionization (SAND and JACOBSEN 1981). It has
been shown that alkalinization of urine (pH>7) results in a decrease in serum
methotrexate concentrations of between 40% and 77% (NIRENBERG et a1.
1977). Alkalinization of urine increases the degree of ionization of this weak
acid, preventing passive reabsorption and hence increasing its renal clearance.
However, SAND and JACOBSEN (1981) interpreted their findings in terms of
186 A. SOMOGYI
increasing the solubility of methotrexate in urine, although this was not di-
rectly assessed.
~) Indomethacin
There has been to systematic pharmacokinetic study investigating the interac-
tion with this nonsteroidal anti-inflammatory drug. However, there have been
case reports of acute renal failure and death, particularly in elderly patients
taking indomethacin and methotrexate (ELLISON and SERVI 1985). It is unclear
whether the mechanism involves competition between indomethacin and
methotrexate for active renal tubular secretion or, more likely, in the elderly
Drug Interactions Involving Renal Excretory Mechanisms 187
y) Ketoprofen
Although described as a pharmacokinetic study, details were lacking in a
retrospective study in 36 patients who were on a number of cycles of high-dose
(mean 3200mg/m2) methotrexate. Four of the nine patients who developed
severe toxicity were also taking ketoprofen and three of these patients died.
There was a substantial increase in the serum methotrexate concentration in
those patients with toxicity (THYSS et al. 1986).
8) Naproxen
In a comprehensive study, 15 patients aged between 30 and 78 years with
rheumatoid arthritis received oral and intravenous methotrexate 15 mg with
and without naproxen at a dose of 1000mg/day for 26 days (STEWART et al.
1990b). Twelve of the patients completed all four arms of the study and, for the
three who did not complete the study, withdrawal was not associated with drug
treatment. There was no significant difference in the total body clearance of
methotrexate with or without naproxen (103 ± 35 alone and 113 ± 48ml/min
with naproxen). In addition, the renal clearance of methotrexate from either
routes (oral and intravenous) was not altered by naproxen. It should be noted
that these patients had normal creatinine clearance values (80mllmin per
1.73m2). TRACY et al. (1992) showed significant (P < 0.05) but small decreases
in the total body clearance of methotrexate (168 ± 50-131 ± 42 ml/min) in nine
patients taking naproxen 750-1250mg/day, with no statistically significant
difference in renal clearance (117 ± 35 mllmin methotrexate alone and 96 ±
38ml/min with naproxen). AHERN et al. (1988) also found no difference in
plasma methotrexate concentrations in three patients with and without con-
comitant naproxen dosing. There has, however, been a report of serious tox-
icity in patients receiving methotrexate and naproxen (SINGH et al. 1986).
£) Ibuprofen
In nine patients with rheumatoid arthritis, the addition of ibuprofen 2.4-
3.6 g/day significantly (P < 0.05) reduced the total and renal clearances of
methotrexate from 168 ± 50 to 101 ± 37mllmin and from 117 ± 35 to 70 ± 27 mIl
min, respectively (TRACY et al. 1992).
Other non-steroidal anti-inflammatory drugs such as diclofenac, diflunisal,
flurbiprofen, sulindac and azapropazone have been investigated, albeit usually
with small numbers (n = 1-3) of patients. Hence, data interpretation has not
allowed conclusions to be deduced, although methotrexate toxicity occurred
in some patients. The interaction between methotrexate and non-steroidal
188 A. SOMOGYI
1. Cimetidine
Cimetidine has been shown to reduce the renal clearance of the following
organic cations: procainamide and its active metabolite n-acetylprocainamide,
triamterene, ranitidine, amiloride, metformin, pindolol, nicotine and
quinidine.
a) Procainamideln-Acetylprocainamide
Several studies have been conducted in healthy subjects and patients and case
reports have highlighted the clinical importance of this interaction.
In the original study (SOMOGYI et al. 1983), six healthy subjects took 1 g
procainamide orally with and without 400mg cimetidine (1 h before the
procainamide dose and 200mg 4-hourly for 12h thereafter) with blood and
urine samples collected over 12 and 48h, respectively. Cimetidine significantly
(P < 0.025) increased the area under the plasma concentration-time curve of
procainamide by an average of 44% and prolonged the half-life from an
harmonic mean of 2.92 to 3.68h (Fig. 3). This was due to a marked reduction
(P < 0.025) in the renal clearance of procainamide from 347 ± 46 to 196 ± 11 mll
min (Fig. 4). In the same study, the area under the plasma concentration-time
curve for n-acetylprocainamide was significantly (P < 0.025) increased by an
average of 25% with a commensurate reduction (P < 0.025) in renal clearance
from 258 ± 60 to 197 ± 49mllmin. Similar data were obtained by CHRISTIAN et
Drug Interactions Involving Renal Excretory Mechanisms 189
100()
5·0
E
......
CI
~
--...
I:
.S! 1·0
CU
I:
CII
U
I:
0
U 0·5
CU
E/I)
.!
Q.
al. (1984) in nine healthy subjects in which cimetidine increased the area under
the plasma concentration-time curve of procainamide by 40% (P < 0.0005),
prolonged its half-life by 24% and reduced its renal clearance by 36% (P <
0.0005). In addition, the area under the plasma concentration-time curve of n-
acetylprocainamide increased (P < 0.03) by 26% but there was no change in
renal clearance. In seven healthy Chinese subjects administered separate
single oral doses of 200mg and 400mg cimetidine (LAI et al. 1988), the area
under the plasma concentration-time curves of procainamide (500mg oral
dose) was increased by 24% (P < 0.01) and 38% (P < 0.05), respectively, with
significant (P < 0.01) reductions in renal clearance of 31 % and 41 %, respec-
tively. These data reinforced the mechanism of competitive inhibition of tubu-
190 A. SOMOGYI
10-12
Time Periods (h)
Fig. 4. Mean ± SD 2 hourly renal clearance values of procainamide when given alone
as 19 procainamide to six healthy subjects (open bars) and when co-administered with
cimetidine (hatched bars). The reduction in renal clearance by cimetidine was signifi-
cant (P < 0.02). (From SOMOGYI et al. 1983)
lar secretion of procainamide by cimetidine. Only the higher dose (400 mg) of
cimetidine significantly increased (mean 45%) the area under the plasma
concentration-time curve and reduced the renal clearance (16%) of n-acetyl-
procainamide. In a steady-state pharmacokinetic study in six healthy young
male subjects who received 500mg sustained release procainamide every 6h
for 13 doses, cimetidine 1200mg/day for 4 days significantly (P < 0.01) in-
creased the procainamide area under the plasma concentration-time curve by
a mean of 43% and significantly (P < 0.01) decreased renal clearance by a
mean of 34 % (RODVOLD et al. 1987). Steady-state serum n-acetylprocainamide
concentrations were significantly increased (P < 0.001) by a mean of 42%. The
results of these four studies in healthy subjects are similar and all groups
proposed the mechanism to involve inhibition of the active tubular secretions
of procainamide and n-acetylprocainamide by cimetidine via competitive
inhibition for the organic cation transporter. The interaction has been de-
scribed and mechanism verified in vitro (McKINNEY and SPEEG 1982). In a
study in 36 hospitalized male patients (aged 65-90 years) who were receiving
oral sustained release procainamide every 6 h, the addition of cimetidine
300mg every 6h for 3 days resulted in a significant (P < 0.005) increase in the
steady-state serum concentrations of procainamide (by a mean of 55%), and
n-acetylprocainamide (mean 36%) (BAUER et al. 1990). In ten of the patients
from whom urine was also collected, the ratio of pro cain amide renal clearance
to creatinine renal clearance was reduced by cimetidine by an average of 33 %
from 2.1 ± 0.6 to 1.4 ± 0.4 (P < 0.02). The n-acetylprocainamide to creatinine
clearance ratio also significantly (P < 0.05) declined from 1.4 ± 0.4 to 1.1 ± 0.4.
Twelve of these patients experienced mild to severe symptoms relating to
procainamide toxicity. Nine of the patients had symptoms of nausea, weak-
ness, malaise and PR interval increases of <20%. Three patients had severe
procainamide toxicity from prolongation of the PR interval of >25%, unifocal
premature ventricular contractions of <30/h and ventricular rates> 150/min for
Drug Interactions Involving Renal Excretory Mechanisms 191
greater than 30s. These latter patients had pro cain amide serum concentra-
tions of greater than 14mg/l and n-acetylprocainamide concentrations of
greater than 16mg/l. All patients with adverse effects had cimetidine discon-
tinued, and procainamide was also discontinued in those patients with severe
toxicity. The procainamide adverse effects disappeared within 24h after cessa-
tion of cimetidine therapy. In a case report (HIGBEE et al. 1984), a 71-year-old
male patient was being treated with procainamide (937.5mg every 6h) for
frequent multiform premature ventricular depolarizations and had achieved
steady-state therapeutic serum procainamide and n-acetylprocainamide con-
centrations of 9 and 7 mg/l, respectively. Following the addition of cimetidine
300mg every 6h for a benign gastric ulcer, the patient was re-admitted with
increased fatigue, weakness, nausea, anorexia and urinary retention. Examina-
tion revealed congestive heart failure and the ECG demonstrated sinus
rhythm with first-degree A V block accompanied by a decreased heart rate.
The procainamide and n-acetylprocainamide serum concentrations were both
15 mg/l, in the range associated with toxicity. The dose of procainamide was
subsequently decreased to 750mg every 6h and procainamide and n-
acetylprocainamide serum concentrations declined to 10 and 12mg/l, respec-
tively. Other dosage adjustments were made but the authors concluded by
stating that their case report should alert clinicians to the potentially danger-
ous adverse effects of this combination (cimetidine and procainamide) as well
as the need to evaluate carefully the source of gastrointestinal symptoms
which may be due either to ulcer disease or procainamide toxicity. They also
commented that the changes encountered suggested that older people may be
at a greater risk of pro cain amide toxicity when prescribed cimetidine.
b) Ranitidine
In a study in six male subjects, the administration of cimetidine 400mg 12
hourly resulted in a significant (P < 0.05) increase in the area under the plasma
ranitidine concentration-time curve from a single 150-mg oral dose of
ranitidine. This was associated with a significant reduction in the renal clear-
ance of ranitidine from 326 ± 67 to 244 ± 57 mllmin (VAN CRUGTEN et al. 1986).
c) Triamterene
In a similar design to the above study (MUIRHEAD et al. 1986), cimetidine
significantly (P < 0.03) increased the area underneath the plasma triamterene
concentration-time curve during a 24-h dosing interval of 100mg daily
triamterene. This was associated with a reduction in the renal clearance of
triamterene from 71 ± 46 to 21 ± 14ml/min. However, the decrease in renal
triamterene clearance occurred only during the first 6 h of the triamterene
dosing interval, at the time of maximum plasma cimetidine concentrations.
When the unbound fraction (0.4) of triamterene in plasma is taken into consid-
eration, the renal clearance due to tubular secretion (minus that due to
reabsorption) was reduced by cimetidine from a mean of 22mllmin to minus
192 A. SOMOGYI
28mllmin. The latter figure indicates that cimetidine completely blocked the
renal tubular secretion of triamterene and uncovered significant triamterene
passive reabsorption, a mechanism previously reported in vitro in the rat (KAu
1978). Cimetidine had no effect on the renal clearance of the major active
metabolite of triamterene, the sulphate conjugate of p-hydroxytriamterene
(MUIRHEAD et al. 1986).
d) Metformin
Cimetidine significantly (P < 0.01) increased the area under the plasma
metformin concentration-time curve by an average of 50% in seven young
healthy subjects given a 0.25-g daily dose of metformin and a 400-mg twice
daily dose of cimetidine. The increase in steady-state plasma metformin con-
centrations was associated with a significant (P < 0.01) reduction (mean 27 % )
in the renal clearance of metformin from 527 ± 165 ml/min to 378 ± 165 ml/min
(SOMOGYI et al. 1987). The reduction in metformin renal clearance only oc-
curred over the first 6h of cimetidine dosing, a phenomenon consistent with
competitive inhibition of metformin renal tubular secretion.
e) Pindolol
In a study to examine the potential stereoselectivity of the inhibition of renal
tubular secretion of organic bases by cimetidine, eight healthy young subjects
received a single 15-mg oral dose of racemic pindolol with 400mg cimetidine
taken twice daily for 2 days (SOMOGYI et al. 1992). The area under the plasma
concentration-time curve for R-(+) pindolol was significantly (P < 0.01) in-
creased by cimetidine from 230 ± 90 to 344 ± 78 ng/ml x h and for S-(-) pin dolo I
it was increased from 209 ± 73 to 288 ± 69ng/ml x h (P < 0.01). Cimetidine
significantly reduced the renal clearance of R-(+) pindolol from 170 ± 55mll
min to 104 ± 88ml/min (P < 0.01) and of S-(-) pindolol from 222 ± 66mllmin
to 155 ± 38mllmin (P < 0.01). The enantiomer with the higher renal clearance
[S-(-) pindolol] had a smaller (mean 26%) cimetidine-induced reduction
in renal clearance compared with R-(+) pindolol (mean 34%) (Fig. 5).
Cimetidine has a stereoselective inhibitory effect on the active transport of
organic cations in the proximal tubule.
f) Disopyramide
In a study in seven healthy male subjects, co-administration of cimetidine
400mg twice daily did not alter the disposition or the renal clearance of the
enantiomers of disopyramide when given as an intravenous bolus of 150mg
racemic disopyramide (BONDE et al. 1991). The reason for this lack of effect of
cimetidine on the renal clearance of a drug (an organic cation) which under-
goes extensive tubular secretion (renal clearance of unbound enantiomers of
disopyramide is 200-400mllmin) is not known. However, since the drug exhib-
its significant concentration-dependent plasma binding, the unbound concen-
trations need to be measured at all blood sampling times and not just at one
Drug Interactions Involving Renal Excretory Mechanisms 193
-s:::
E
:::;:
300
250 #
•
0
1m
R-(+)-pindolol
S-(-)-pindolol
-
R-(+)-pindolol + cimetidine
E
~ S-(-)-pindolol + cimetidine
-...
s::: 200
0
CD 150
u
CD
CJ) 100
...as
50
::s
.0
.-
::s 0
Fig. 5. Net renal clearance via tubular secretion for R-(+) pindolol and S-(-) pindolol
when racemic pindolol was administered alone (15 mg) and when co-administered with
cimetidine in eight subjects. Values represent mean ± SD. *P < 0.001, enantiomer alone
vs. enantiomer + cimetidine; #p < 0.001, R-(+) pindolol vs. S-(-) pindolol. (From
SOMOGYI et al. 1992)
particular time (or concentration) point. As was the case with triamterene and
metformin, the effect of cimetidine may only occur in the first few hours after
dosing, particularly with the low dose of cimetidine used_ Hence to observe an
effect on the renal clearance of disopyramide may have required frequent
urine collections and not reliance solely placed on urine collected over a long
time intervaL
g) Amiloride
In eight healthy subjects given a single dose of 5 mg amiloride, cimetidine
400 mg twice daily reduced the renal clearance of amiloride by an average of
17% from 358 ± 134 to 299 ± 118mllmin (P < 0.05), but there was no effect on
the area under the plasma amiloride concentration-time curve (SOMOGYI et aL
1989). The effect on renal amiloride clearance was small in magnitude and
occurred only in the 2- to 4-h time period after cimetidine dosing.
h) Nicotine
In six healthy subjects, cimetidine 600mg 12 hourly for 2 days significantly
(P < 0_05) reduced the renal clearance of nicotine (as an intravenous infusion
of 1,ug/kg per minute for 30min) from 1.49 ± 0.30 to 0.79 ± 0.30mllmin per
kilogram, whereas there was no effect (P> 0.05) on the renal clearance of the
nicotine metabolite cotinine (0.14 ± 0.04mllmin per kilogram without
cimetidine vs_ 0.15 ± 0.10mllmin per kilogram with cimetidine) (BENDAYAN et
al. 1990). Since the renal clearance of nicotine when co-administered with
cimetidine was below the renal clearance due solely to filtration (fu x GFR),
these results indicate the existence of passive reabsorption of nicotine in
addition to active tubular secretion.
194 A. SOMOGYI
i) Quinidine
In six healthy subjects, HARDY et al. (1983) showed that cimetidine given as
1.2g/day reduced the oral clearance of quinidine (as 400mg quinidine
sulphate) by 37% (P < 0.05) with no change in the urinary excretion of
unchanged quinidine. As a consequence, calculation of the renal clearance
allows one to conclude that cimetidine reduced the renal clearance of quini-
dine from 5.6 to 3.611h.
j) CephalothiniCephalexin
In order to investigate the specificity of cimetidine-induced reductions in the
renal tubular secretion of organic cations, VAN CRUGTEN et al. (1986) showed,
in six normal healthy subjects, that cimetidine (400mg twice daily) had no
effect (P> 0.05) on the renal clearance of the organic anion cephalothin (305
± 127mllmin alone vs. 284 ± 141mllmin with cimetidine), whose renal clear-
ance is inhibited by probenecid (Table 1). However, for the zwitterion cephal-
exin, cimetidine reduced (P < 0.01) the renal clearance from 263 ± 39 to 208 ±
21 mllmin. The renal clearance of this zwitterion, which has both acidic (pKa
2.6) and basic (pKa 7.0) functional groups, is also reduced by probenecid
(Table 1). These investigators concluded that cimetidine's effect was specific
for organic cations, and that, for those zwitterions that undergo tubular secre-
tion, both organic anion and cation transport systems might be involved.
2. Ranitidine
In the light of the inhibitory effect of cimetidine on the renal tubular secretion
of other organic cations, studies in humans have determined whether other Hz
antagonists (which are organic cations) behave in a similar manner to
cimetidine.
a) Procainamideln-Acetylprocainamide
In six healthy subjects, the administration of 150mg twice daily ranitidine l3h
prior to a 1-g oral procainamide dose increased the area under the plasma
procainamide concentration-time curve by an average of 14% with a signifi-
cant (P < 0.05) but small (mean 18%) reduction in the renal clearance of
procainamide (SOMOGYI and BOCHNER 1984). The area under the plasma n-
acetylprocainamide concentration-time curve was also reduced by an average
of 11 % (P < 0.05). In the same study, when the dose of ranitidine was increased
to 750mg in three subjects, the area under the plasma concentration-time
curve for procainamide was increased by an average of 20% and renal clear-
ance was reduced by an average of 35%, and for n-acetylprocainamide, renal
clearance was reduced by an average of 38%. This study demonstrated that in
the clinical doses used, ranitidine will have little inhibitory effect; however, if
larger doses are used or if the patient's renal function declines such that in
both cases higher plasma ranitidine concentrations are achieved, then inhibi-
Drug Interactions Involving Renal Excretory Mechanisms 195
b) Triamterene
In eight healthy male subjects, ranitidine 150mg twice daily reduced the renal
clearance of triamterene (100mg daily for 4 days) by a mean of 51 % (P < 0.05)
from 94 ± 64 to 46 ± 42mllmin (MUIRHEAD et al. 1988). This was interpreted as
competition between triamterene and ranitidine for the renal organic cation
transport system. In the same study, the renal clearance of the sulphate conju-
gate of p-hydroxytriamterene was significantly (P < 0.01) reduced by ranitidine
from 105 ± 35 to 56 ± 20 mllmin. There was no effect of ranitidine on the degree
of binding of the sulphate conjugate to plasma components (fu = 0.09). Al-
though the results for triamterene are consistent with inhibition of tubular
secretion of triamterene by another organic cation (ranitidine), the inhibition
of the renal clearance of the sulphate conjugate raised the possibility of multi-
transport systems, as cimetidine which reduced triamterene renal clearance
did not alter the renal clearance of the sulphate conjugate (see Sect. C.IL1.c).
c) Nicotine
In six healthy subjects, ranitidine 300mg 12 hourly for 2 days had no effect on
the renal clearance of nicotine (intravenous infusion of 1 Jig/kg per minute for
30min) (1.49 ± 0.30 alone and 0.88 ± 0.40mllmin per kilogram with ranitidine)
or cotinine (BENDAYAN et al. 1990).
3. Famotidine
a) Procainamideln-Acetylprocainamide
Eight healthy subjects were given a single 5-mg/kg intravenous infusion of
procainamide alone and after 5 days of receiving 40mg famotidine once daily
(KLOTZ et al. 1985). Famotidine had no significant effect (P> 0.05) on the total
body clearance or renal clearance of procainamide and n-acetylprocainamide.
Although famotidine has a high renal clearance of more than 300ml/min, its
half-life is less than 3 h; hence plasma concentrations from a daily 40-mg dose
may have been insufficient to elicit inhibitory effects on the tubular secretion
of procainamide and n-acetylprocainamide. In addition, it has been observed
196 A. SOMOGYI
that the organic anion probenecid inhibits the tubular secretion of famotidine
(see Sect. c.I.l.l).
4. Trimethoprim
a) Procainamideln-Acetylprocainamide
In eight healthy subjects, procainamide 500mg orally every 6h for 3 days was
co-administered with and without trimethoprim 200mg daily for 4 days
(KOSOGLOU et al. 1988). Trimethoprim significantly (P < 0.05) increased the
steady-state plasma concentrations of procainamide by an average of 62 % and
for n-acetylprocainamide by 52%. These were due to significant (P < 0.05)
reductions in the renal clearances of procainamide (mean 47%) and n-
acetylprocainamide (mean 13%). These investigators detected small but
statistically significant (P < 0.0001) increases in the QTc interval when
trimethoprim was co-administered, presumably as a result of the elevated
plasma concentrations of procainamide and n-acetylprocainamide. The same
investigators also conducted a study in ten healthy young male subjects who
received a single l-g oral procainamide dose alone and with trimethoprim
100mg twice daily for 2 days prior to and 200mg with the procainamide dose
(VLASSES et al. 1989). The area under the plasma procainamide concentration-
time curve was significantly (P < 0.05) increased by trimethoprim by an aver-
age of 39% and the renal clearance was reduced from 487 ± 129 to 267 ±
123ml/min (P < 0.05). For n-acetylprocainamide, the area under the plasma
concentration-time curve was increased by an average of 27% by tri-
methoprim (P < 0.05) and renal clearance was reduced from 275 ± 78 to
192 ± 82mllmin (P < 0.05). Since trimethoprim is actively taken up by the
renal cortex via the organic cation transport system, and has a high renal
clearance (>220mllmin for the unbound drug), this interaction is consistent
with trimethoprim having a higher affinity than procainamide and n-
acetylprocainamide for the organic cation transporter and hence inhibiting
their tubular secretion.
b) Zidovudine
In nine HIV-infected patients with normal renal function, the effect of
trimethoprim (150mg orally) on the disposition of zidovudine (3mg/kg intra-
venous infusion) was evaluated (CHAlTON et al. 1992). Trimethoprim signifi-
cantly (P < 0.05) reduced the renal clearance of zidovudine from 0.34 ± 0.15 to
0.17 ± 0.091/h per kilogram, an average decrease of 58%. In addition, the
apparent renal clearance of the glucuronide conjugate of zidovudine was also
significantly (P < 0.05) reduced by trimethoprim from 0.70 ± 0.21 to 0.56 ±
0.1211h per kilogram. In the same study but on another occasion, the patients
received a single dose of trimethoprim 150 mg plus sulphamethoxazole 800 mg.
The decrease in zidovudine renal clearance averaged 48% and for the glucu-
ronide conjugate 20%, similar to when trimethoprim was administered alone,
Drug Interactions Involving Renal Excretory Mechanisms 197
5. Amiodarone
a) Procainamideln-Acetyiprocainamide
The disposition of procainamide was investigated in eight patients who
received an intravenous dose of between 6 and 15 mg/kg before and after
1-2 weeks treatment with amiodarone (1.6g1day) (WINDLE et al. 1987).
Amiodarone significantly (P < 0.01) reduced the total body clearance of
procainamide from 0.43 ± 0.12 to 0.33 ± 0.12l/kg per hour. In two of the
patients, urine was also collected and amiodarone reduced the renal clearance
of pro cain amide from 0.25 and 0.241/kg per hour to 0.14 and 0.151/kg per hour,
respectively. Hence the reduction in total body clearance appears to be pre-
dominantly due to renal clearance, most likely a consequence of competition
for active tubular secretion. Since the half-life of amiodarone is almost 1
month, the degree of inhibition of procainamide renal tubular secretion might
be even higher when steady-state plasma concentrations of amiodarone are
reached after about 4 months of chronic dosing.
6. Quinine/Quinidine
a) Amantadine
In a study designed to evaluate the effect of age and gender on inhibition of
amantadine renal clearance by quinine and quinidine (GAUDRY et al. 1993), a
comparison was made between nine young 27- to 39-year-old subjects (five
male and four female) and nine 60- to 72-year-old adults (four male and five
female). Amantadine (an organic base undergoing tubular secretion) was
administered as a 3-mglkg oral syrup on the evening before the study and
quinine (as quinine sulphate 200mg) and quinidine (as quinidine sulphate
200mg) were administered orally approximately 12h later. Blood and urine
samples were collected at 2, 4, 6 and 8h after quinine/quinidine dosing and all
urine was collected in 2 hourly aliquots to 8h. Quinine and quinidine had no
significant (P> 0.05) effect on amantadine renal clearance in either young or
old subjects. However, when the data were separated according to gender,
quinine and quinidine significantly (P < 0.05) reduced the amantadine renal
clearance in males but not in females. When both male and female data were
combined, only quinine reduced the renal clearance of amantadine. The inves-
tigators cited animal studies which suggest that testosterone may enhance the
renal organic anion and cation transport of xenobiotics. However, it is unclear
as to how this would impact on inhibition of transport as there was no gender
198 A. SOMOGYI
1. Digoxin
Digoxin is the most commonly used cardiac glycoside drug for the treatment of
congestive heart failure and atrial fibrillation. When administered by the intra-
venous route, approximately 60% of the dose is recovered unchanged in urine
and its renal clearance is in excess of the clearance via glomerular filtration
(taking into account the degree of plasma binding), indicating that this chemi-
cally neutral compound undergoes tubular secretion (STEINESS 1974). A large
number of interactions have been reported for digoxin and several of these
involve renal excretory mechanisms.
a) Amiodarone
Six patients with either ventricular or supraventricular tachyarrhythmias
were given a 1-mg intravenous dose of digoxin alone and after 2 weeks of
amiodarone 1600mg/day (NADEMANEE et al. 1984). The renal clearance of
digoxin was reduced by an average of 22% (from 0.85 ± 0.33 to 0.66 ± 0.39ml/
min per kilogram) but this was not significant (P> 0.05). However, in ten
healthy subjects who were administered 400 mg amiodarone daily for 3 weeks,
there was a mean 22% decrease (from 105 ± 39 to 84 ± 15mllmin; P < 0.05) in
the renal clearance of digoxin when administered as a 1-mg intravenous dose
(FENSTER et al. 1985). Amiodarone also reduced the non-renal clearance of
digoxin in both studies. The reduction in renal clearance was interpreted as a
reduction in the tubular secretion of digoxin as there was no effect on the renal
clearance of creatinine by amiodarone (NADEMANEE et al. 1984).
digoxin is secreted (KOREN 1987). It has been reported that amiloride at a dose
of 5mg twice daily for 8 days in six healthy subjects increased (P < 0.001)
digoxin renal clearance from 1.3 ± 0.1 to 204 ± 0.5ml/min per kilogram follow-
ing a 15-.ug/kg intravenous bolus dose of digoxin without altering inulin clear-
ance (WALDORFF et al. 1981). The mechanism by which amiloride increases the
renal clearance of digoxin was attributed to increased potassium in distal
tubular cells and therefore increasing digoxin secretion, although this latter
mechanism was not proven.
c) Quinidine
Several mechanisms, including a reduction in digoxin renal clearance, are
involved in the interaction of quinidine on digoxin disposition. The reduc-
tion in the renal clearance of digoxin by quinidine has been reported as: (a) a
mean of 35% reduction in three patients on long-term oral digoxin and admin-
istered 250mg twice daily quinidine, with no effect on creatinine clearance
(HOOYMANS and MERKUS 1978); (b) a mean of 33% reduction (from 1.64 ± 0.60
to 1.09 ± 0.24mllmin per kilogram; P < 0.05) in six patients given a 1-mg
intravenous bolus dose of digoxin with and without 200mg quinidine 6 hourly
for 7 days (HAGER et al. 1979); (c) a mean of 34% reduction (from 53 ± 21 to
35 ± 13mllmin per 1.73m2 ; P < 0.001) in 15 patients prescribed between 0.125
and 0.25mg/day digoxin and 200-300mg/day quinidine, with no alteration in
creatinine clearance (MUNGALL et al. 1980); (d) a mean of 52% decrease (from
130 ± 32 to 62 ± 10ml/min; P < 0.01) in digoxin renal clearance in five patients
given an intravenous dose of tritiated digoxin with and without quinidine 0.6 g
twice daily (SCHENCK-GUSTAFSSON and DAHLQVIST 1981); (e) a decrease of
32% in mean renal clearance of digoxin (from 1.93 ± 0.61 to 1.32 ± Oo4Oml/min
per kilogram) in seven healthy subjects receiving quinidine 800mg/day and
increasing to a mean 54% reduction in digoxin renal clearance (from 1.47 ±
0.30 to 0.68 ± 0.22; P < 0.001) when the dose of quinidine was doubled to
1600mg/day for 4 days (LEAHEY et al. 1981). Digoxin was administered as a
1-mg intravenous infusion over 60min on both occasions in both groups. A
relationship (r = 0.60) was found between the serum quinidine concentration
and reduction in digoxin renal clearance. Thus, the reduction in renal clear-
ance of digoxin depends on the concentration of quinidine and is consistent
with competition for active tubular secretion; (f) In a comprehensive study in
eight healthy young male subjects, digoxin 0.5-0.75mglday was given alone
and with 400mg twice daily quinidine (in four subjects). The renal clearance of
digoxin was significantly reduced (P < 0.05) by an average of 29% from 155 ±
40 to 110 ± 21 mllmin. In the other four subjects, the effect of quinine (the
diastereoisomer of quinidine) given as 750 mg/day was investigated. The renal
clearance of digoxin was not altered by quinine (177 ± 40 ml/min digoxin alone
and 185 ± 53 ml/min with quinine) (HEDMAN et al. 1990).
Since quinidine has no effect on glomerular filtration rate, the results of
these studies have been interpreted as a reduction in the tubular secretion of
200 A. SOMOGYI
digoxin by quinidine. It is possible that both digoxin and quinidine compete for
the same carrier-mediated transport system in the kidney. Quinidine competes
for the organic cation transporter; however, it has been shown in animals that
digoxin is not eliminated by this transport system (KOREN 1987). Clearly
further studies are warranted to delineate the mechanism of the renal tubular
secretion of digoxin.
d) Verapamil
Verapamil has been shown to increase plasma digoxin concentrations and
produce toxicity and in some cases fatality (as reviewed by STOCKLEY 1994). In
eight healthy subjects who received a l-mg intravenous infusion of digoxin
over 15 min, the addition of verapamil 800mg orally three times daily for 10
days resulted in a reduction in the renal clearance of digoxin from 2.2 ± 0.4 to
1.7 ± 0.5mllmin per kilogram (P < 0.025) (PEDERSEN et al. 1981). Similarly, in
seven patients receiving verapamil (80mg three times daily) for long-term
therapy, the renal clearance of digoxin from a dose of 125 J.lglday was reduced
after 1 week of verapamil therapy by an average of 36% (from 198 ± 46 to 128
± 27ml/min; P < 0.001), however, after 6 weeks of verapamil there was no
reduction in the renal clearance of digoxin (203 ± 71 mllmin) (PEDERSEN et al.
1982). In a study by BELZ et al. (1983) in healthy subjects who were adminis-
tered digoxin 0.375mglday to steady state, the addition of verapamil (80mg
three times/day for 2 weeks) reduced (P < 0.05) the digoxin renal clearance
from 218 ± 90 to 148 ± 42mllmin per 1.73m2 • In a more recent study in six
patients with chronic atrial fibrillation, verapamil (240mg/day) had no effect
on the renal clearance of digoxin (dose varied between 0.25 and O.5mg/day)
(HEDMAN et al. 1991). The renal clearance of digoxin was 153 ± 51mllmin
when given alone and 173 ± 51 ml/min when given with verapamil (P > 0.05).
This study found a significant association between the renal clearance of
digoxin and urine flow rate (r = 0.75; P < 0.001). At a urine flow rate of lmll
min the renal clearance of digoxin was about 100mllmin and at 4mllmin the
renal clearance had doubled to almost 200mllmin. There was no difference in
urine flow rate between the two study occasions. It would appear that the
effect of verapamil on digoxin renal clearance may be transient and may be
confounded by alterations in urine flow rate.
The mechanism for the renal tubular secretion of digoxin remains un-
known. Studies in animals have indicated that digoxin is not secreted by the
organic cation or anion transport system. There was also no evidence to
indicate that binding to the antiluminal Na+/K+-ATPase alters the renal clear-
ance of digoxin (KOREN 1987).
D. Tubular Reabsorption
1. Lithium
Lithium is used for the management of manic depressive psychoses, and has a
relatively narrow therapeutic index in which toxicity is manifested by impaired
consciousness, disorientation, ataxia, tremor, muscle twitches and vomiting.
These adverse effects usually occur at plasma concentrations greater than
2mmolll. The kidney is the major clearance organ for the elimination of
lithium. After filtration of lithium and sodium at the glomerulus, approxi-
mately 70% of the filtered load of sodium and lithium is reabsorbed at the
proximal tubule, which does not distinguish between these two inorganic ions.
Thus, the renal clearance of lithium is similar and parallels that of sodium and
the ratio of the renal clearance of lithium to that of creatinine is approximately
0.2. A number of drugs alter the clearance of lithium through altering its renal
clearance. These include non-steroidal anti-inflammatory drugs and thiazide
diuretics, which reduce the renal clearance of lithium, and sodium salts and
theophylline, which increase the renal clearance of lithium.
a) Theophylline
In ten healthy subjects with normal renal function administered lithium car-
bonate 900mg daily for 7 days, the addition of theophylline to achieve steady-
state plasma concentrations of between 5 and 12mg/1 significantly (P < 0.05)
increased the renal clearance of lithium by an average of 42% from 18.3 ± 7.0
to 26.1 ± 11.5mllmin and shortened the half-life from 28.1 ± 7.5 to 21.8 ± 6.6h
(PERRY et al. 1984). There was also a significant decrease in serum lithium
concentrations by an average of 30%. This interaction was dependent on the
plasma theophylline concentration, as there was a statistically significant (P <
0.05) linear relationship between the plasma theophylline concentration and
the percentage change in lithium clearance. The incidence of side effects
(restlessness, tremor and anorexia) has been reported to be significantly
greater when theophylline was co-administered to patients (COOK et al. 1985).
In a single case report of a patient with mania treated with lithium, the
addition of theophylline resulted in a rapid onset of relapse of mania. When
the dosage of lithium was increased, serum lithium concentrations increased
202 A. SOMOGYI
and the patient's mania was controlled (SIERLES and OSSOWSKI 1982). The
mechanism for the increase in the renal clearance of lithium by theophylline is
unknown. Theophylline does not alter renal blood flow or glomerular filtration
rate and therefore appears to reduce the tubular reabsorption of lithium.
b) Sodium Salts
THOMSEN and SCHOU (1968) originally reported that administration of sodium
bicarbonate with lithium resulted in a 30% increase in the ratio of lithium
renal clearance to creatinine clearance, and DEMERS and HENINGER (1971)
reported in six patients with mania, receiving lithium 1200-1800mg/day, that
when a high-sodium diet (120-250mmol/day) was compared to a low-sodium
(69-91 mmoI/day) diet, there was a decline in the serum lithium concentration
from a mean of 0.99 to 0.83 mmol/l, and an increase in manic effects and
behaviour ratings. In a case report, a male with depressive illness was pre-
scribed 250 mg lithium carbonate four times daily to achieve a target lithium
serum concentration of 0.5 mmol/I. When the frequency of dosage was in-
creased to six times a day his serum lithium concentration was still below
0.6mmoI/1 as the patient was also taking sodium bicarbonate. When this was
ceased, serum lithium concentrations reached 0.8mmol/1 on the initial dosage
(ARTHUR 1975). In a subsequent case study, MCSWIGGAN (1978) reported on
several patients who failed to achieve and maintain therapeutic serum lithium
concentrations. It was subsequently revealed that a nurse had been adminis-
tering a proprietary saline drink for upset stomachs, which contained about
50% sodium bicarbonate. When this was ceased, two patients developed
lithium toxicity. Other studies have shown similar effects in that intake of
sodium salts reduces serum lithium concentrations, resulting in a reduced
pharmacodynamic effect of lithium (DEMERS and HENINGER 1971).
The mechanism for this interaction is consistent with the known mecha-
nism for the renal elimination of lithium. When plasma sodium concentrations
are elevated through the addition of sodium-containing preparations, extracel-
lular fluid is expanded and the kidney reduces its reabsorption of sodium and,
as a consequence, lithium, resulting in increased renal clearance and reduced
plasma concentrations. This interaction is clinically important and has implica-
tions for the management of mania in patients.
c) Diuretics
a) Thiazide Diuretics
There has been no systematic pharmacokinetic study investigating the interac-
tion between lithium and thiazide diuretics. Nevertheless, there have been a
number of clinically important case reports, which have highlighted a poten-
tially serious interaction between these drugs in which thiazide diuretics in-
crease serum lithium concentrations. For example, in 22 patients receiving
hydroflumethiazide or bendrofluazide, the renal clearance of lithium was sig-
Drug Interactions Involving Renal Excretory Mechanisms 203
~) Loop Diuretics
There have been no comprehensive studies investigating potential interactions
between loop diuretics and lithium, although there have been a number of case
reports and some studies in normal healthy subjects. In five normal healthy
subjects administered 300mg lithium three times daily, resulting in steady-
state serum concentrations of 0.4 mmolll, addition of 40 mg frusemide daily for
14 days produced no lithium-induced adverse effects or increase in serum
lithium concentrations in five of six subjects. However, one subject withdrew
from the study because of toxicity which was associated with a 61 % increase
in serum lithium concentration (from 0.44 to 0.71 mmol/l; JEFFERSON and
KALIN 1979). In a case report bumetanide has also been associated with the
development of lithium toxicity in a patient (KERRY et al. 1980), in which the
serum lithium concentration increased from between 0.8 and 1.1mmol/l to
1.6mmol/l.
The mechanism for this interaction is likely to be similar to that described
for the thiazide diuretics.
a) Indomethacin
Although published in abstract from (LEFTWICH et al. 1978), it was reported
that in five healthy subjects taking 300-600mg lithium carbonate daily, addi-
tion of indomethacin 50mg three times a day significantly reduced the renal
clearance of lithium by an average of 31 % with a significant (P < 0.05) 43%
increase in serum lithium concentrations. The urinary excretion of the me-
tabolite of prostaglandin (PGE z) synthesis was reduced by indomethacin. In
ten normal healthy female subjects on a balanced sodium diet, addition of
150mg indomethacin daily to lithium sulphate (330mg twice daily) signifi-
cantly increased the serum lithium concentration from 0.68 ± 0.07 mmol/l to
0.84 ± O.13mmol/l (REIMANN et al. 1983). This was accompanied by a signifi-
cant decrease in the renal clearance of lithium (from 26 ± 7 mIlmin to 19 ± 5 mIl
min; P < 0.001) and urinary PGE z excretion was reduced from 298 ± 46 to 168
± 89ng/24h (P < 0.005). Reports of lithium toxicity associated with increased
serum lithium concentrations in patients also taking indomethacin have been
described (RAGHEB et al. 1980).
~) Ibuprofen
In a study in 11 healthy young (23-38 years) subjects, ibuprofen 1600mg daily
increased the serum lithium concentration (resulting from a dose of 450mg
lithium sulphate twice daily) by an average of 15%, from 0.67 ± 0.12 to 0.77 ±
O.13mmoIlI (P < 0.001) (KRISTOFF et al. 1986), with a significant decrease in
renal clearance (24 ± 5 to 19 ± 4mIlmin, n = 5). In patients maintained on
lithium, the addition of 1800mg ibuprofen increased serum lithium concentra-
tions by between 12% and 66% (RAGHEB 1987a). There have been reports of
lithium toxicity in patients receiving ibuprofen (KHAN 1991).
y) Diclofenac
Five young healthy women, on a standardized sodium diet of 150mmoIlday,
were administered lithium as a sustained release tablet of 330mg twice daily.
Diclofenac 50mg three times daily was also administered (on another occa-
sion) and it significantly (P = 0.002) decreased the renal lithium clearance by an
average of 23% and significantly (P < 0.001) increased serum lithium concen-
trations by a mean of26%. Renal prostaglandin synthesis was measured by the
urinary excretion of PGEz and was decreased by 53% (mean) compared with
the control period (213 ± 81 to 103 ± 36 ng/24 h) (REIMANN and FROLICH 1981).
0) Naproxen
In seven patients stabilized on lithium, the addition of 750mg naproxen re-
sulted in a 16% (range 0%-42%) increase in serum lithium concentrations
(from 0.81 ± 0.11 to 0.94 ± 0.08mmol/l). The renal clearance of lithium de-
creased from 17 ± 5 to 13 ± 4mllmin (P < 0.05) (RAGHEB and POWELL 1986).
One patient developed signs of toxicity and had an increase in the serum
lithium concentration from 0.95 to 1.13 mmoIll.
Drug Interactions Involving Renal Excretory Mechanisms 205
€) Piroxicam
A case report documented an increase in serum lithium concentrations in a
patient also taking piroxicam which resulted in adverse effects (KERRY et al.
1983).
~) Phenylbutazone
Although no systematic pharmacokinetic study has been conducted, there has
been a case report in which, in six patients with a bipolar affective disorder,
treatment with 300mg phenylbutazone caused a small increase (range 0%-
15%) in serum lithium concentrations, which was associated in some patients
with side effects (RAGHEB 1990).
11) Sulindac
In six patients requiring lithium therapy at doses ranging from 600 to 900mgl
day, the addition of sulindac 150mg twice a day did not significantly (P> 0.05)
alter serum lithium concentrations (0.84 ± 0.07 lithium alone vs. 0.83 ±
O.lOmmolll with sulindac) (RAGHEB and POWELL 1986).
glandin synthesis. The renal sparing effect of sulindac would be consistent with
its lack of interaction with lithium. This interaction is clinically important as a
number of reports of lithium toxicity due to non-steroidal anti-inflammatory
drugs excluding sulindac and aspirin/salicylate have been documented.
a) Chlorpropamide
Six healthy subjects received a 250-mg oral dose of chlorpropamide on
several occasions, once when urine pH was made acidic (pH 4.7-5.5) with
ammonium chloride and once when alkaline (pH 7.1-8.2) with sodium bicar-
bonate (NEUVONEN and KARKKAINEN 1983). The area under the plasma
chlorpropamide concentration-time curve was significantly (P < 0.001) larger
when urine was acidic (3635 ± 1212mg/1 x h) than when alkaline (700 ± 98mg/
I x h), and there were clear differences in half-life from a mean of 13h under
alkaline urine conditions to 69 h when urine was acidic. These substantial
differences were attributed to a marked dependence of renal clearance on
urine pH. When all the data were combined, providing 144 sets of urine pH
and renal clearance data, it was noted that, at a urine pH of 5, renal clearance
was 1 mllh but, when urine pH was 7, renal clearance was 100mllh, a two
orders of magnitude difference. This is probably the best example of how urine
pH can influence the disposition of a drug. For chlorpropamide, an acidic,
lipophilic drug with a pKa of 4.8, by raising urine pH, the degree of ionization
is increased thus preventing passive reabsorption and increasing apparent
renal clearance; in contrast, when urine pH is acidic, the degree of unioniza-
tion is increased, resulting in increased passive reabsorption and negligible
renal clearance (0.017 mllmin).
b) Pseudoephedrine
In eight subjects, who received a 5-mg/kg oral dose of pseudoephedrine,
BRATER et al. (1980) found a strong relationship between the half-life of
pseudoephedrine (a surrogate marker of renal clearance) and urine pH, such
that at pH 5.8 the half-life was about 300min and at pH 7.2 it was 600min.
These investigators also found that for each subject, when urine was alkaline
(pH >7), there was a significant correlation (r = 0.71-0.99) between renal
clearance and urine flow. Thus for this lipophilic organic cation with a pKa of
9.4, its renal clearance is highly dependent on both urine pH and flow rate. The
Drug Interactions Involving Renal Excretory Mechanisms 207
Acknowledgement. This review has been supported by the National Health and Medi-
cal Research Council of Australia.
References
Ahern M, Booth J, Loxton A, McCarthy P, Meffin P, Kevat S (1988) Methotrexate
kinetics in rheumatoid arthritis: is there an interaction with nonsteroidal anti-
inflammatory drugs? J RheumatoI15:1356-1360
Aherne GW, Piall E, Marks V, Mould G, White WF (1978) Prolongation and enhance-
ment of serum methotrexate concentrations by probenecid. Br Med J 1:1097-1099
Andreasen F, Sigurd B, Steiness E (1980) Effect of probenecid on excretion and
natriuretic action of furosemide. Eur J Clin Pharmacol 18:489--495
Arthur RK (1975) Lithium levels and "Soda BIC". Med J Aust 2:918
Baber N, Halliday L, Sibeon R, Littler T, Orme M L'E (1978) The interaction between
indomethacin and probenecid. Clin Pharmacol Ther 24:298-307
Bauer LA, Black D, Gensler A (1990) Procainamide-cimetidine drug interaction in
elderly male patients. J Am Geriatr Soc 38:467-469
Belz GG, Doering W, Munkes R, Matthews J (1983) Interaction between digoxin and
calcium antagonists and antiarrhythmic drugs. Clin Pharmacol Ther 33:410-417
Bendayan R, Sullivan JT, Shaw C, Frecker RC, Sellers EM (1990) Effect of cimetidine
and ranitidine on the hepatic and renal elimination of nicotine in humans. Eur J
Clin PharmacoI38:165-169
Beyer KJ, Russo HF, Tillson EK, Miller AK, Verwey WF, Gass SR (1951) "Benemid",
p-(di-n-propylsulfamyl)-benzoic acid: its renal affinity and its elimination. Am J
Physiol 166:625-640
Bloom EJ, Ignoffo RJ, Reis CA, Cadman E (1986) Delayed clearance (CL) of
methotrexate (MTX) associated with antibiotics and antiinflammatory agents.
Clin Res 34:560A
Bonde J, Pedersen LE, Nygaard E, Ramsing T, Angelo HR, Kampmann JP (1991)
Stereos elective pharmacokinetics of disopyramide and interaction with
cimetidine. Br J Clin Pharmacol 31:708-710
Borga 0, Larsson R, Lunell E (1986) Effects of probenecid on enprofylline kinetics in
man. Eur J Clin Pharmacol 30:221-223
Bourke RS, Cheda G, Bremer A, Watanabe 0, Tower DB (1975) Inhibition of renal
tubular transport of methotrexate by probenecid. Cancer Res 35:110-116
Brater DC, Kaojarern S, Benet LZ, Lin ET, Lockwood T, Morris RC, McSherry EJ,
Melmon KL (1980) Renal excretion of pseudoephedrine. Clin Pharmacol Ther
28:690-694
Brown GR (1993) Cephalosporin-probenecid drug interactions. Clin Pharmacokinet
24:289-300
Burnell JM, Kirby WMM (1951) Effectiveness of a new compound, benemid, in elevat-
ing serum penicillin concentrations. J Clin Invest 30:697-700
Chatton JY, Munafo A, Chave JP, Steinhauslin F, Roch-Rame1 F, Glauser MP, Biollaz
J (1992) Trimethoprim, alone or in combination with sulphamethoxazole, de-
creases the renal tubular secretion of zidovudine and its glucuronide. Br J Clin
PharmacoI34:551-554
Christian CD, Meredith CG, Speeg KV (1984) Cimetidine inhibits renal pro cain amide
clearance. Clin Pharmacol Ther 36:221-227
Cook BL, Smith RE, Perry PJ, Calloway RA (1985) Theophylline-lithium interaction.
J Clin Psychiatry 46:278-279
208 A. SOMOGYI
Corvaia L, Li SC, Ioannides-Demos LL, Bowes G, Spicer WJ, Spelman DW, Tong N,
McLean AJ (1992) A prospective study of the effects of oral probenecid on the
pharmacokinetics of intravenous ticarcillin in patients with cystic fibrosis. J
Antimicrob Chemother 30:875-878
De Miranda P, Good SS, Yarchoan R, Thomas RV, Blum MR, Myers CE, Broder S
(1989) Alteration of zidovudine pharmacokinetics by probenecid in patients with
AIDS or AIDS-related complex. Clin Pharmacol Ther 46:494-500
Demers RG, Heninger GR (1971) Sodium intake and lithium treatment in mania. Am
J Psychiatry 128:132-136
Elion GB, Yii T-F, Gutman AB, Hitchings GH (1968) Renal clearance of oxipurinol,
the chief metabolite of allopurinol. Am J Med 45:69-77
Ellison NM, Servi RJ (1985) Acute renal failure and death following sequential inter-
mediate-dose methotrexate and 5-FU: a possible adverse effect due to concomi-
tant indomethacin administration. Cancer Treat Rep 69:342-343
Fenster PE, White NW, Hanson CD (1985) Pharmacokinetic evaluation of the digoxin-
amiodarone interaction. J Am Coll Cardiol5:108-112
Furst DE, Herman RA, Koehnke R, Ericksen N, Hash L, Riggs CE, Porras A, Veng-
Pedersen P (1990) Effect of aspirin and sulindac on methotrexate clearance.
J Pharm Sci 79:782-786
Gaudry SE, Sitar DS, Smyth DD, McKenzie JK, Aoki FY (1993) Gender and age as
factors in the inhibition of renal clearance of amantadine by quinine and quinidine.
Clin Pharmacol Ther 54:23-27
Gilman AG, Rall TW, Nies AS, Taylor P (eds) (1990) The pharmacological basis of
therapeutics, 8th edn. Pergamon, New York
Hager WD, Fenster P, Mayersohn M, Perrier D, Graves P, Marcus FI, Goldman S
(1979) Digoxin-quinidine interaction. N Engl J Med 300:1238-1241
Hardy BG, Zador IT, Golden L, Lalka D, Schentag JJ (1983) Effect of cimetidine on
the pharmacokinetics and pharmacodynamics of quinidine. Am J Cardiol52:172-
175
Hedaya MS, Elmquist WF, Sawchuk RJ (1990) Probenecid inhibits the metabolic
and renal clearances of zidovudine (AZT) in human volunteers. Pharm Res 7:411-
417
Hedman A, Angelin B, Arvidsson A, Dahlqvist R, Nilsson B (1990) Interactions in the
renal and biliary elimination of digoxin: stereoselective difference between qui-
nine and quinidine. Clin Pharmacol Ther 47:20-26
Hedman A, Angelin B, Arvidsson A, Beck 0, Dahlqvist R, Nilsson B, Olsson M,
Schenck-Gustafsson K (1991) Digoxin-verapamil interaction: reduction of biliary
but not renal digoxin clearance in humans. Clin Pharmacol Ther 49:256-262
Hedman A, Angelin B, Arvidsson A, Dahlqvist R (1992) Digoxin-interactions in man:
spironolactone reduces renal but not biliary digoxin clearance. Eur J Clin
Pharmacol 42:481-485
Hendel J, Nyfors A (1984) Nonlinear renal elimination kinetics of methotrexate due to
saturation of renal tubular reabsorption. Eur J Clin Pharmacol26:121-124
Higbee MD, Wood JS, Mead RA (1984) Procainamide-cimetidine interaction: a poten-
tial toxic interaction in the elderly. J Am Geriatr Soc 32:162-164
Himmelhoch JM, Poust RI, Mallinger AG, Hanin I, Neil JF (1977) Adjustment of
lithium dose during lithium-chlorothiazide therapy. Clin Pharmacol Ther 22:225-
227
Homeida M, Roberts C, Branch RA (1977) Influence of probenecid and
spironolactone on furosemide kinetics and dynamics in man. Clin Pharmacol Ther
22:402-409
HonariJ, Blair AD, Cutler RE (1977) Effects of probenecid on furosemide kinetics and
natriuresis in man. Clin Pharmacol Ther 22:395-401
Hooymans PM, Merkus FWMH (1978) Effect of quinidine on plasma concentration of
digoxin. Br Med J 2:1022
Howell SB, Olshen RA, Rice JA (1979) Effect of probenecid on cerebrospinal fluid
methotrexate kinetics. Clin Pharmacol Ther 26:641-646
Drug Interactions Involving Renal Excretory Mechanisms 209
A. Introduction
Pharmacodynamic interactions occur at receptor sites or result more broadly
from the effect of the drugs on physiological processes. Both antagonism and
synergism may be produced. The synergistic interactions, as far as they have
the potential to be beneficial and clinically useful, are reviewed separately in
this handbook (see Chap. 8).
The present chapter will concentrate on adverse drug interactions result-
ing in either a diminished therapeutic effect or an increased toxicity. Its aim is
to describe the mechanisms by which pharmacodynamic interactions may
occur and to illustrate them with clinically relevant examples. It is not a
compilation of all possible and known interactions, duplicating standard
publications (SCHOU 1982; MciNNES and BRODIE 1988; STOCKLEY 1991;
BUGNON et al. 1993; HANSTEN et al. 1993). Accordingly, the classification
adopted here is based on sites of action and not on drug classes. Different steps
in the same process, e.g., synaptic transmission, or different mechanisms tak-
ing part in the regulation of one system or function, e.g., blood pressure, may
be affected.
Since they are based on mechanisms of action supposedly known to
the prescriptor, pharmacodynamic interactions should be more easily pre-
vented and should have less clinical impact than pharmacokinetic interactions.
However, the relevance of such a statement may be questioned in view of
the recent advances in the characterization and detection of kinetic
interactions.
I. Transmitter Systems
1. Noradrenergic Synapse
The various drugs interfering with practically every step of the nor adrenergic
transmission represent a well-established source of pharmacodynamic interac-
tions (Fig. 1). In the CNS, where adrenaline is a neurotransmitter on its own
216 M. SCHORDERET and J.D. FERRERO
(MEFFORD 1988; COOPER et al. 1991), similar interactions can occur. The spe-
cific plasma membrane noradrenaline transporter has been cloned and charac-
terized (PACHOLCZYK et al. 1991; USDIN et al. 1991). It plays a role in the
mechanism of action and potential interactions of amphetamine and tyramine,
which stimulate the outward transport of noradrenaline, as well as of cocaine
and imipramine, which block the inward transport (reuptake) of noradrena-
line (Fig. 1).
Possible interactions at postsynaptic receptors are not shown in Fig. 1,
since they can easily be anticipated. Patients on ,B-blockers are particularly at
risk (Table 1). For example, the blockade of ,B-receptors makes a patient
resistant to adrenaline in the emergency treatment of anaphylaxis. In this
TYROSINE L-DOPA
+
DOPAMINE
+
NORADRENALINE
i:~::\
~ ~
~G\!Y
/RES,
--e--- MAO[-A
• • • COC AMPH/TYR
• • ~
2. Dopaminergic Synapse
Dopamine is a neurotransmitter in the eNS, and as a circulating catechola-
mine released from the adrenals it also plays multiple roles in the periphery
(HORN and MURPHY 1991). In addition, brain dopamine receptors are the
primary target in the treatment of schizophrenia, Parkinson's disease and
Huntington's chorea (SEEMAN and VAN TOL 1994). A better knowledge of the
processes involved in the dopaminergic transmission, as well as of the pharma-
cological characteristics of the subtypes of receptors (D! to D s), may help to
understand and possibly to avoid drug-drug interactions (Fig. 2).
The interaction between L-dopa and pharmacological doses of pyridoxine
(vitamin B 6 ) is due to an increased peripheral decarboxylation of L-dopa by
+
(VITAMIN 86)
DOPAMINE
t
RES
MAOI- B
COC AMPH/TYR
3. Serotonergic Synapse
The last decade has been marked by a rapid progress in serotonin (5-hydroxy-
tryptamine, 5-HT) research with the cloning, expression and characterization
of numerous subtypes of 5-HT receptors (BOESS and MARTIN 1994), as well as
of the specific plasma membrane transporter of serotonin (BLAKELY et al. 1991;
HOFFMAN et al. 1991).
The serotonin transporter is concerned with the mechanism of action
and possible interactions of amphetamine and 3,4-methylenedioxy-
methamphetamine (MDMA, "ecstasy"), which stimulate the outward trans-
port of serotonin, as well as of cocaine and the selective serotonin reuptake
inhibitors (SSRls), which block the inward transport of serotonin (Fig. 3). The
combined treatment with irreversible MAOls and SSRls has been shown to
cause a serotonin syndrome (FEIGHNER et al. 1990; GRAM 1994; SPINA and
PERUCCA 1994). Moreover, a lethal serotonin syndrome has been reported
after the concomitant administration of high-dose moclobemide with
citalopram or clomipramine (NEUVONEN et al. 1993; MITCHELL 1994). Thus, the
Drug-Drug Interactions at Receptors and Other Active Sites 221
TRYPTOPHAN _ _ _ +- 5-HYDROXY -
TRYPTOPHAN
+
5-HYDROXYTRYPT AMINE
(5 - HT)
(SEROTONIN)
•• COC AMPH/MDMA
~
prospective benefits of combining SSRIs with MAOIs - even with the revers-
ible MAO I-A moclobemide - in the treatment of depression should be seri-
ously weighed against the associated risks (SPINA and PERUCCA 1994).
Like cocaine, the OTC antitussive dextromethorphan, as well as pethidine
(meperidine), can also block the neuronal uptake of serotonin (BROWNE and
LINTER 1987; STACK et al. 1988; HILL et al. 1992; CETARUK and AARON 1994).
The association of dextromethorphan or pethidine with MAOIs (including
222 M. SCHORDERET and J.D. FERRERO
4. Cholinergic Synapse
In the cholinergic transmission, unlike the monoaminergic transmissions,
drugs affecting the synthesis (e.g., choline, lecithins), storage and uptake of the
ACET)
CHOLINE
~~r--- UPTAKE
RELEASE
INHIBITORS
NON DEPOLARIZING • • •
NEUROMUSCULAR • •
BLOCKING DRUGS
CHOLINESTERASE
INHIBITORS
transmitter have not been found therapeutically useful. A few drugs, however,
have been shown to interfere presynaptically with acetylcholine release at
the neuromuscular junction (Fig. 4). Apart from the botulinum toxins, some
antibiotics (aminoglycosides, polymyxins, lincosamides) (SINGH et al. 1982),
quinidine, quinine and procainamide, as well as magnesium salts, playa role in
this effect (CLARKE adn MIRAKHUR 1994). The aminoglycosides are not only
able to produce weakness on their own or aggravate the muscular symptoms in
myasthenia gravis (DRACHMAN 1994), they may also enhance the effect of the
neuromuscular blocking agents (CLARKE and MIRAKHUR 1994).
Most interactions occurring at cholinergic receptors involve additive
effects between muscarinic receptor blocking drugs, examples of which can be
found in many therapeutic classes (Table 2). In the nicotinic domain, additive
effects with dangerous cardiovascular consequences have also been reported
in patients treated with transdermal nicotine delivery systems who do not stop
smoking or who start smoking again (FIORE et al. 1992).
Cholinesterase inhibitors are routinely used in anaesthesiology to reverse
curare-induced neuromuscular block (Fig. 4). As the use of cholinesterase
inhibitors, notably tacrine, has been proposed in the treatment of Alzheimer's
disease, it should be remembered that any agent with antimuscarinic proper-
ties which crosses the blood-brain barrier will interact negatively. A more
insidious interaction of anticholinergic drugs is the inhibition of the
gastrokinetic effect of the 5-HT4 agonists metoclopramide and cisapride that
act by stimulating acetylcholine release in the enteric nervous system
(SCHOURKES et al. 1988).
5. GABAergic Synapse
GAB A is the main inhibitory transmitter in the mammalian brain. In postsyn-
aptic neurons, it produces either a fast response, through the ligand-gated
ion-channel GABAAreceptor, or a slow modulatory response, through the G-
protein-coupled GABAB receptor (MODY et al. 1994). As shown in Table 3,
various substances can either facilitate or inhibit the GABAA-mediated
increase in CI- conductance (SIEGHART 1992).
224 M. SCHORDERET and J.D. FERRERO
Benzodiazepines Flumazenila
Barbiturates
Gabapentin Penicillins
Progabide Quinolones
Vigabatrin
Ethanol Bicuculline
General anaesthetics Picrotoxin
A vermectin insecticides Cyclodiene insecticides
sodium channel in the resting state and the affinity increases when the channel
is in the activated (quinidine) or inactivated (lidocaine) states. Class I agents
are further characterized by the association/dissociation kinetics to and from
the channel receptor site (WOOSLEY 1991).
The concomitant administration of antiarrhythmic drugs with distinct
properties (e.g., class Ia/lb) may result in either adverse or beneficial interac-
tions (LANGIERI et al. 1995). As an example of the latter, mexiletine (Ib) and
quinidine (Ia) in combination have been found to be more effective at a lower
dose and to produce fewer adverse effects than each drug given alone in higher
dosage (BIGGER 1984; GIARDIA and WESCHSLER 1990). This may be explained
by the fact that quinidine-induced prolongation of repolarization prolongs
the inactivated state of the sodium channel. As a result, the binding of
mexiletine, which possesses higher affinity for the channel in the inactivated
state, is increased. Moreover, the competition between "fast" and "slow"
sodium channel blockers (including non-antiarrhythmic drugs) may be
effective in reversing toxic electrophysiologic effects. Lidocaine has been
shown to reverse the QRS prolongation due to cocaine (BAUMAN et al. 1994)
or propoxyphene (WHITCOMB et al. 1989) overdoses. Finally, the drugs that
cause repolarization abnormalities (see Sect. B.II.2) tend to have greater
toxicity at slower heart rate. Increasing heart rate by temporary pacing or
isoprenaline infusion is the recommended treatment in this case (BEN-DAVID
and ZIPES 1993).
Cardiac calcium channels, particularly in the atrioventricular node, are
concerned with a well-known additive interaction between class II (f3-
blockers) and class IV (verapamil, diltiazem) antiarrhythmic drugs (see also
Table 1). The resulting inhibition of atrioventricular conduction is due to the
blockade of nodal calcium channels induced both directly by the calcium
channel blocker and indirectly, through a reduction in sympathetic drive by
the f3-blocker (WINNIFORD et al. 1982).
Antiarrhythmic drugs
- Class Ia Quinidine
- Class III Sotalol
Antihypertensive agents Ketanserine
Antimalarial drugs Chloroquine, mefioquine, halofantrine
CNS stimulants 3,4-Methylenedioxy-methamphetamine ("ecstasy")
Diuretics Hydrochlorothiazide
Hj-antihistaminics Terfenadine, astemizole
Macrolide antibiotics Erythromycin
Neuroleptics Chlorpromazine, haloperidol
Tricyclic antidepressants Imipramine
like terfenadine and astemizole, can create a life-threatening risk when admin-
istered in overdose or in association with macrolide antibiotics, particularly
erythromycin. In the latter case, the interaction is largely of a pharmacokinetic
nature (see Chap. 6, this volume).
1. Adrenal Corticosteroids
Interactions implying the corticosteroids can be deduced from their broad
spectrum of activity. Their metabolic effects, which tend to produce a
Drug-Drug Interactions at Receptors and Other Active Sites 227
hyperglycaemic state, will reduce the efficacy of insulin and other antidiabetic
agents (see below). Those glucocorticoids possessing mineralocorticoid activ-
ity will aggravate the hypokalaemia induced by thiazide or loop diuretics.
Finally, the corticosteroids may interfere with the development of the protec-
tive immune response to vaccines (BUTLER and ROSSEN 1977; ZIELENSKI et al.
1986). Moreover, in view of altered defense mechanisms, live vaccines should
not be administered to patients treated with systemic corticosteroids (GROSS et
al. 1985).
2. Glycaemic Regulation
The association of insulin and oral hypoglycaemic agents has been advocated
in the treatment of type II diabetes (GERICH 1989; LEWITT et al. 1989). Clearly
enough, the additive effects of such a combination would increase the risk of
severe hypoglycaemia. Similarly, the a-glucosidase inhibitors (e.g., acarbose)
may aggravate insulin- and sulphonylurea-induced hypoglycaemia (BALFOUR
and McTAVISH 1993). Inversely, the therapeutic efficacy of oral antidiabetic
agents would be reduced by the simultaneous administration of thiazide
diuretics or of diazoxide (see Sect. B.I1.3).
Many drugs affect glycaemic regulation and can thus interfere with
antidiabetic agents. It has been mentioned previously (see Sect. B.Ll) that
f3-blockers may mask the early signs of hypoglycaemia and should be avoided
in diabetic patients. In addition, non-selective f3-blockers have been reported
to cause severe hypoglycaemia in both diabetic and non-diabetic patients
(KOTLER et al. 1966; ANGELO-NIELSON 1980). Unexpectedly, the salicylates in
Hyperglycaemia Hypoglycaemia
f3-Blockers f3-Blockers
A-Adrenergic agonists
Cyclosporine Disopyramide
Corticosteroids Angiotensin-converting enzyme
inhibitors (ACEIs)
Diazoxide Ethanol
Nicotinic acid Fibric acid derivatives
Octreotide
Pentamidine Pentamidine
Phenothiazines Quinine
Phenytoin Salicylates
Thiazide diuretics Streptozotocin
Thyroid hormones
high doses (4-6g1day) have been shown to decrease blood glucose concentra-
tions (SELTZER 1989). Salicylates increase insulin secretion in non-insulin-
dependent diabetes, increase insulin sensitivity and inhibit lipolysis (MICOSSI
et al. 1978; PANDIT et al. 1993). They can thus enhance the hypoglycaemic
effects of insulin and oral antidiabetic agents (RICHARDSON et al. 1986). Addi-
tional pharmacokinetic interactions with the sulphonylureas (e.g., displace-
ment from protein-binding sites) may also playa role (see Chap. 4, this
volume). If anti-inflammatory doses of aspirin are to be prescribed to a dia-
betic patient, they should be started low and increased progressively with
frequent blood glucose monitoring.
As mentioned in Sect. B.II!.l, the glucocorticoids represent another
cause of interaction with antidiabetic agents. By stimulating hepatic gluconeo-
genesis and decreasing peripheral utilization of glucose (JACKSON and
BRESSLER 1981; GERICH 1989), they tend to produce a diabetes-like state
(PANDIT et al. 1993).
This survey is limited to a few classical examples of pharmacodynamic
interactions that implicate insulin and other antidiabetic agents. Many
additional drugs (including ethanol) can interfere with glycaemic regulation
(Table 5).
PROSTAGLANOINS ANGIOTENSIN n
3. Potassinm Homeostasis
All thiazide and loop diuretics increase the renal excretion of potassium and
reduce kalaemia. Hypokalaemia augments the risk of arrhythmias, particu-
230 M. SCHORDERET and J.D. FERRERO
larly in the course of therapy with antiarrhythmic drugs (see Sect. B.I1.2). It is
also well known to increase the toxicity of cardiac glycosides (STEINESS and
OLESEN 1976). In order to treat hypokalaemia, certain patients will require the
prescription of potassium supplements or of a potassium-sparing diuretic. In
these patients, the co-prescription of ACE inhibitors creates a serious risk of
hyperkalaemia (BURNAKIS and MIODUCH 1984). By interfering with renin re-
lease, the NSAIDs tend to produce hyperkalaemia (TAN et al. 1979), which
would also be aggravated by the simultaneous prescription of potassium-
sparing diuretics or of potassium salts (ZIMRAN et al. 1985).
References
Amrein R, Giintert TW, Dingemanse J, Lorscheid T, Stabl M, Schmid-Burgk W (1992)
Interactions of moclobemide with concomitantly administered medication: evi-
dence from pharmacological and clinical studies. Psychopharmacology (Berl)
106:S24-S31
Angelo-Nielson K (1980) Timolol topically and diabetes mellitus. JAMA 244:263
Balfour JA, McTavish D (1993) Acarbose. An update of its pharmacology and
therapeutic use in diabetes mellitus. Drugs 46:1025-1054
Bauman JL, Grawe JJ, Winecoff AP, Hariman RJ (1994) Cocaine-related sudden
cardiac death: a hypothesis correlating basic science and clinical observations. J
Clin Pharmacol 34:902-911
Ben-David J, Zipes DP (1993) Torsades de pointes and proarrhythmia. Lancet
341:1578-1582
Berlin I, Zimmer R, Cournot A, Payan C, Peclariosse AM, Puech AJ (1989) Determi-
nation and comparison of the pressor effect of tyramine during long-term
moclobemide and tranylcypromine treatment in healthy volunteers. Clin
Pharmacol Ther 46:344-351
Bigger JT (1984) The interaction of mexiletine with other cardiovascular drugs. Am
Heart J 107:1079-1085
Blackwell B, Marley E (1966) Interactions of cheese and its constituents with monoam-
ine oxidase inhibitors. Br J Pharmacol Chemother 26:120-141
Blakely RD, Berson HE, Fremeau RT, Caron MG, Peek MM et al. (1991) Cloning
and expression of a functional serotonin transporter from rat brain. Nature
354:66-70
Bochner BS, Lichstenstein LM (1991) Anaphylaxis. N Engl J Med 324:1785-1790
Boess FG, Martin IL (1994) Molecular biology of 5-HT receptors. Neuropharmacology
33:275-317
Browne B, Linter S (1987) Monoamine oxidase inhibitors and narcotic analgesics. Br J
Psychiatry 151:210-212
Bugnon 0, Dommer J, Mesnil M, Schorderet M (1993) Detection and management of
drug interactions (in French). Swiss Society of Pharmacy, Bern (Basic formation,
vol 1)
Burnakis TG, Mioduch HJ (1984) Combined therapy with captopril and potassium
supplementation. A potential for hyperkalemia. Arch Intern Med 144:2371-2372
Butler WT, Rossen RD (1977) Effects of corticosteroids on immunity in man. I.
Decreased serum IgG concentration caused by 3 or 5 days of high doses of
methylprednisolone. J Clin Invest 30:1451-1455
Cetaruk EW, Aaron CK (1994) Hazards of nonprescription medications. Emerg Med
Clin North Am 12:483-510
Christ W, Lehnert T, Ulbrich B (1988) Specific toxicologic aspects of the quinolones.
Rev Infect Dis 10 [Suppl1]:S141-S146
Clarke RSJ, Mirakhur RK (1994) Adverse effects of muscle relaxants. Adv Drug React
Toxicol Rev 13:23-41
Drug-Drug Interactions at Receptors and Other Active Sites 231
Cooper JR, Bloom FE, Roth RH (1991) The biochemical basis of neuropharmacology,
6th edn. Oxford University Press, New York
Curtis DR, Game CJA, Johnston GAR, McCulloch RM, MacLachlan RM (1972)
Convulsive action of penicillin. Brain Res 43:242-245
Dilsaver SC (1993) Antipsychotic agents: a review. Am Fam Physician 47:199-
204
Drachman DB (1994) Myasthenia gravis. N Engl J Med 330:1797-1809
Drug and Therapeutics Bulletin (1992) Sumatriptan: a new approach to migraine. Drug
Ther Bull 30:85-87
Edwards G, Weston AH (1993) The pharmacology of ATP-sensitive potassium chan-
nels. Annu Rev Pharmacol Toxicol 33:597-637
Favre L, Glasson P, Riondel A, Vallotton MB (1983) Interaction of diuretics and non-
steroidal anti-inflammatory drugs in man. Clin Sci 64:407-415
Feighner JP, Boyer WF, Tyler DL, Neborsky RJ (1990) Adverse consequences of
fluoxetine-MAOI combination therapy. J Clin Psychiatry 51:222-225
Fiore MC, Jorenby DE, Baker TB, Kenford SL (1992) Tobacco dependence and the
nicotine patch; clinical guidelines for effective use. JAMA 268:2687-2694
Fitton A, Faulds D, Goa KL (1992) Moclobemide. A review of its pharmacological
properties and therapeutic use in depressive illness. Drugs 43:561-596
Gerich JE (1989) Oral hypoglycemic agents. N Engl J Med 321:1231-1245
Giardia E, Weschsler M (1990) Low dose quinidine-mexiletine combination therapy
versus quinidine monotherapy for treatment of ventricular arrhythmias. J Am Coli
Cardiol 15:1138-1145
Gieschke R, Schmid-Burgk W, Amrein R (1988) Interaction of moclobemide, a new
reversible monoamine oxidase inhibitor with oral tyramine. J Neural Transm
[Suppl 26]:97-104
Giros B, Mestikawy L, Bertrand L, Caron MG (1991) Cloning and functional
characterization of a cocaine-sensitive dopamine transporter. FEBS Lett 295:149-
154
Glassman AH, Bigger JT Jr (1981) Cardiovascular effects of therapeutic doses of
tricyclic antidepressants. Arch Gen Psychiatry 38:815-820
Gram LF (1994) Fluoxetine. N Engl J Med 331:1354-1361
Gross P A, Gould A, Brown AE (1985) Effect of cancer chemotherapy on the immune
response to influenza virus vaccine: review of published studies. Rev Infect Dis
7:613-618
Guy EJ (1992) Selective serotonin reuptake inhibitors. Br Med J 304:1655-1658
Halliwell RF, Davey PG, Lambert 11 (1993) Antagonism of GABAA receptors by 4-
quinolones. J Antimicrob Chemother 31:457-462
Hansten PD, Horn JR, Koda-Kimble MA, Young LY (eds) (1993) Drug interactions
and updates quarterly. Applied Therapeutics, Vancouver
Hill S, Yau K, Whitwam J (1992) MAOIs to RIMAs in anaesthesia - a literature
review. Psychopharmacology (Berl) 106 [Suppl]:S43-S45
Hoffman BJ, Mezby E, Brownstein MJ (1991) Cloning of a serotonin transporter
affected by antidepressants. Science 254:79-80
Horn PT, Murphy MB (1991) Dopamine receptor agonists in cardiovascular medicine.
Trends Cardiovasc Med 1:103-107
Hricik DE, Dunn MJ (1990) Angiotensin-converting enzyme inhibitor-induced
renal failure: causes, consequences, and diagnostic uses. J Am Soc Nephrol1:845-
858
Jackson EJ, Bressler R (1981) Clinical pharmacology of sulphonylurea hypoglycemic
agents. Drugs 22:211-245, 295-320
Johnson AG, Nguyen TV, Day RO (1994) Do nonsteroidal anti-inflammatory drugs
affect blood pressure? A meta-analysis. Ann Intern Med 121:289-300
Kilty JE, Lorang D, Amara SG (1991) Cloning and expression of a cocaine-sensitive rat
dopamine transporter. Science 254:578-579
Kotler MN, Berman L, Rubenstein AH (1966) Hypoglycaemia precipitated by pro-
pranolol. Lancet 11:1389-1390
232 M. SCHORDERET and J.D. FERRERO
Lange RA, Cigarro RG, Flores ED, McBride W, Kim AS, Bedotto JB, Danziger RS,
Hillis LD (1990) Potentiation of cocaine-induced coronary vasoconstriction by
beta-adrenergic blockade. Ann Intern Med 324:1485-1490
Langieri G, Marinchak RA, Rials SJ, Kowey PR (1995) Antiarrhythmic drug interac-
tions. Curr Op in Cardiol 10:26-28
Lejoyeux M, Ades J, Rouillon F (1994) Serotonin syndrome. Incidence, symptoms and
treatment. CNS Drugs 2:132-143
Lewitt MS, Yu VK, Rennie GC, Carter IN, Marel GM, Yue DK, Hooper MJ (1989)
Effects of combined insulin-sulfonylurea therapy in type II patients. Diabetes
Care 12:379-383
Marchant B, Wray R, Leach A, Nama M (1989) Flumazenil causing convulsions and
ventricular tachycardia. Br Med J 299:860
Mars H (1974) Levodopa, carbidopa and pyridoxine in Parkinson's disease. Arch
NeuroI30:444-447
McInnes GT, Brodie MJ (1988) Drug interactions that matter. A critical reappraisal.
Drugs 36:83-110
Mefford IN (1988) Epinephrine in mammalian brain. Prog Neuropsychopharmacol
Bioi Psychiatry 12:365-388
Michell JR, Cavanaugh JH, Arias L, Oates JA (1970) Guanethidine and related agents.
III. Antagonism by drugs which inhibit the norepinephrine pump in man. J Clin
Invest 49:1596-1604
Micossi P, Pontiroli AE, Baron SH, Tamayo RC, Lengel F, Bevilacqua M, Raggi U,
Norbiato G, Foa PP (1978) Aspirin stimulates insulin and glucagon secretion and
increases glucose tolerance in normal and diabetic subjects. Diabetes 27:1196-
1204
Mitchell PB (1994) Selective serotonin reuptake inhibitors: adverse effects, toxicity and
interactions. Adv Drug React Toxicol Rev 13:121-144
Mitchelson F (1992) Pharmacological agents affecting emesis; a review (part I). Drugs
43:295-315
Mody I, De Koninck Y, Otis TS, Soltesz I (1994) Bridging the cleft at GABA synapses
in the brain. Trends Neurosci 17:517-525
Mott DD, Lewis DV (1994) The pharmacology and function of central GABAB recep-
tors. Int Rev Neurobiol 36:97-223
Motwani JG, Struthers AD (1994) Interactive effects of indomethacin, angiotensin II
and frusemide on renal haemodynamics and natriuresis in man. Br J Clin
PharmacoI37:355-361
Neuvonen PJ, Pohjola-Sintonen S, Tacke U (1993) Five fatal cases of serotonin syn-
drome after moclobemide-citalopram or moclobemide-clomipramine overdoses.
Lancet 342:1419
Pacholczyk T, Blakely RD, Amara SG (1991) Expression cloning of a cocaine-
and antidepressant-sensitive human noradrenaline transporter. Nature 350:350-
353
Pandit MK, Burke J, Gustafson AB, Minocha A, Peiris AN (1993) Drug-induced
disorders of glucose tolerance. Ann Intern Med 118:529-539
Remick RA (1988) Anticholinergic side effects of tricyclic antidepressants and
their management. Prog Neuropsychopharmacol Bioi Psychiatry 12:225-
231
Richardson T, Foster J, Mawer GE (1986) Enhancement by sodium salicylate of the
blood glucose lowering effect of chlorpropamide-drug interaction or summation of
similar effects? Br J Clin Pharmacol 22:43-48
Schmid-Antomarchi H, De Weille J, Fosset M, Lazdunski M (1987) The receptor for
antidiabetic sulfonylureas controls the activity of the ATP-modulated K+ channel
in insulin-secreting cells. J Bioi Chern 262:15840-15844
Schou JS (1982) Drug interactions at (pharmacodynamically active) receptor sites.
Pharmacol Ther 17:199-210
Schourkes JA, Van Bergen PJ, Van Nueten JM (1988) Prejunctional muscarinic (M)-
Drug-Drug Interactions at Receptors and Other Active Sites 233
A. Introduction: "Mithridatium"
Synergy is derived from the Greek word O'Vv£P}f5s, meaning working together.
In pharmacological terms it has a precise meaning, which is distinct from
interaction between two or more active substances where the interaction may
be on the absorption, distribution, metabolism or excretion of one or more of
the substances. For a synergistic therapeutic effect to occur there need not
necessarily be an interaction of this type but the effect should be to the
patient's therapeutic advantage.
Historically, the seeking for the universal panacea or cure for all ills has
to some extent hinged on the philosophy of empirical polypharmacy. This
can be traced back to philosophies such as that evolved by Mithridates VI,
King of Pontus 133 B.C.-63 B.c. (see GRIFFIN 1994). Pontus abounded in
medicinal plants and Mithridates acquired considerable knowledge of
them. Like every despot of that period he lived in fear of being assassinated
by poisoning, in consequence of which he sought the universal antidote to
all poisons. Mithridates proceeded along a simple line of reasoning. Having
investigated the powers of a number of single ingredients, which he found
to be the antidote to various venoms and poisons individually, he evaluated
them experimentally on condemned criminals. He then compounded all the
effective substances into one antidote hoping thereby to produce universal
protection. A daily dose was taken prophylactically to give the immunity he
sought.
Pliny wrote: "by his unaided efforts Mithridates devised the plan of drink-
ing poison daily after first taking remedies in order to achieve immunity by
sheer habituation. He was the first to discover the various antidotes, one of
which is even known by his name". So effective was Mithridates' formulation
that he tried unsuccessfully to commit suicide by poisoning and finally killed
himself with a "Celtic sword".
Galen writing in the second century A.D. at a time when he was physician
to the Roman Emperor, Marcus Aurelius, refers to "Mithridatium" and a
formulation derived from it by one Andromachus, Nero's physician. It is said
that Andromachus removed some ingredients from Mithridates' formulation
and added others particularly viper's flesh. To this new product he gave the
name "Galene", which means "tranquillity". Galene later became known as
236 J.P. GRIFFIN and P.F. D' ARCY
C. Diuretic Combinations
Commonly prescribed fixed combinations that can be cited as examples of
believed benefit are combined diuretics. The BRITISH NATIONAL FORMULARY
No. 27 (1994) cites over 25 products comprising a loop or thiazide diuretic
combined with either potassium or a potassium-sparing diuretic. The MeRec
Bulletin (MEDICINES RESOURCE CENTRE 1994) asks the question: Are such
combinations necessary? and concludes that for the vast majority of patients
there is no need for such combinations, and that the routine approach should
be to give a thiazide or loop diuretic alone.
D. Co-trimoxazole
In 1948, HITCHINGS et al. demonstrated that nearly all 2,4-diaminopyrimidines
and related compounds inhibited the growth of Lactobacillus casei by virtue of
their properties as folic acid antagonists. Four years later the precise mode of
action was shown to involve inhibition of the enzyme dihydrofolate reductase.
This is the enzyme which catalyses the transformation of folic acid into a form
that can be utilised in the process leading to thymine formation prior to its
incorporation into DNA. It was eventually shown that the degree of inhibition
of the enzyme by different antimetabolites varied according to the species
from which the enzyme was isolated. This led to the development of species-
specific dihydrofolate reductase inhibitors amongst a series of 5-benzyl deriva-
tives of 2,4-diaminopyrimidine. The antimalarial drug pyrimethamine is one
such example. By varying the substituents on the benzene and pyrimidine
rings, it was found that methoxyl groups enhanced the inhibitory potency
against cultures of Proteus vulgaris, whilst not markedly inhibiting the enzyme
derived from mammalian sources.
This discovery was fully exploited with trimethoprim, the trimethoxy
derivative, which is 50000 times more potent as an inhibitor of bacterial
dihydrofolate reductase as it is of human dihydrofolate reductase (see
SNEADER 1985). Trimethoprim was marketed in fixed combination with
sulphamethoxazole as Septrin and Bactrim by Wellcome and Roche, respec-
tively. This combination was based on the fact that sulphonamides prevent the
bacterial synthesis of dihydrofolate. It was theoretically conceived that if a
bacterium was resistant to the sulphonamide it could be hoped that the
dihydrofolate acid reduction process would be inhibited by trimethoprim or
vice versa. The choice of sulphamethoxazole as the sulphonamide in the
238 J.P. GRIFFIN and P.F. D'ARCY
combination was based purely on the fact that it had approximately the same
duration of action as trimethoprim.
The eventual choice of the name co-trimoxazole by the British
Pharmacopoeia Commission gave a form of sanctity to this combination and it
was a sanctity that time has shown to be ill-deserved.
The BRITISH NATIONAL FORMULARY No. 27 states that "trimethoprim alone
can be used for the treatment of urinary and respiratory-tract infections and
for prostatitis, shigellosis and invasive salmonella infections. Side effects are
less than with co-trimoxazole especially in older patients; therefore it should
be used in place of co-trimoxazole for most infections (except Pneumocystis
carinii infections and respiratory infections associated with AIDS)".
The side effects of co-trimoxazole are the same as those of sulphonamides,
e.g., rashes, Stevens-Johnson syndrome, toxic epidermal necrolysis, renal
failure and blood dyscrasias notably bone-marrow depression and
agranulocytosis. In short, the serious side effects of co-trimoxazole are due to
the sulphonamide component which, apart from a possible but dubious syner-
gistic role in Pneumocystis carinii infections, is not serving any useful purpose.
This example of co-trimoxazole is one where a synergistic role was con-
ceived on good theoretical grounds but was shown after some 12 years to be
unnecessary and undesirable and unsafe for many patients, particularly the
elderly. The sulphonamide component contributes little to the efficacy of the
product but causes most of the combination's adverse reactions.
Despite the fact that the undesirability of this fixed combination has been
known for over 15 years and that repeated comments to this effect have been
made in the BRITISH NATIONAL FORMULARY in every issue since 1981, the
product remains widely used by British general practitioners. Some 18 million
prescriptions per year are written in general practice for co-trimoxazole,
and approximately the same number for trimethoprim. However, in hospital
practice, the use of co-trimoxazole has virtually disappeared.
Table 1. Antiepileptic drugs: single therapy by epilepsy type. (After CHADWICK 1993)
Idiopathic
Simple absences Sodium valproate or Benzodiazepines or
ethosuximide lamotrigine
Juvenile myoclonic Sodium valproate Phenobarbitone or
lamotrigine
Awakening time/clonic Sodium valproate Carbamazepine, phenytoin
or lamotrigine
Symptomatic Sodium valproate or Carbamazepine, phenytoin
benzodiazipines or phenobarbitone
Partial epilepsy Carbamazepine or Phenytoin, phenobarbitone
sodium valproate or vigabatrin
Unclassified Sodium val pro ate Lamotrigine
that their therapeutic effects were additive while their individual toxicity was
reduced, but there is no evidence for this." The wording in the BNF No. 27
(1994) is identical.
The continuing high usage of the phenobarbitone/phenytoin combinations
shows how hard it is to break an established but undesirable prescribing
practice. The usage of the new anticonvulsants (e.g., GABA derivatives,
vigabatrin) remains low. The popularity of the ingrained phenobarbitone/
phenytoin combination is, however, made more attractive by the annual low
price for treatment (see Table 2). Downward pressures by Government on
prescribing costs are an undoubted factor in perpetuating the use of two agents
once thought to have a useful synergistic action. Conversely, the use of new
single-therapy regimens is being discouraged by cost.
F. Antitubercular Drugs
The synergistic use of antibiotics and antibacterial agents in the treatment of
tuberculosis is well established. The treatment is in two phases, an "initial
phase" with at least three drugs and a "continuation phase" using two drugs.
The following regimen is recommended by the British Thoracic Society:
Initial Phase. The concurrent use of three antitubercular agents is designed to
reduce the population of viable bacteria as rapidly as possible. The usual
regimen is the daily use of rifampicin, isoniazid and pyrazinamide; ethambutol
may be added if resistant organisms are suspected. Streptomycin is rarely used
in the United Kingdom, although it is an effective agent for use in the "initial
phase" and can be used instead of isoniazid. The "initial phase" of treatment
should last at least 2 months.
Continuous Phase. This should last a minimum of 4 months using a combina-
tion of isoniazid and rifampicin.
240 J.P. GRIFFIN and P.F. D' ARCY
Treatment regimens for Third World countries may well differ from that
recommended in the United Kingdom and will largely depend on the local
pattern of resistance and the cost and availability of antitubercular drugs.
Dosage regimens can be found in the BRITISH NATIONAL FORMULARY and
the current edition should always be consulted.
G. Anti-Leprosy Treatments
For more than 20 years the use of dapsone alone was commonplace but
multidrug regimens have had to be developed due to the emergence of drug-
resistant strains of the leprosy bacillus. Thus, the principle for the use of multi-
drug treatments in leprosy is the same as that of tuberculosis. For therapeutic
purposes, leprosy can be subdivided into, firstly, multibacillary leprosy, for
which a three-drug regimen is recommended using dapsone, rifampicin and
clofazimine; secondly, paucibacillary leprosy, for which a two-drug regimen of
rifampicin and dapsone are recommended. A 2-year period of treatment is
recommended in both grades of severity and advice from the Panel of Leprosy
Opinion from the Department of Health should be sought before initiating
treatment.
revolutionised the treatment of peptic ulcer and virtually eliminated the need
for the heroic gastric surgery conducted in earlier decades.
Treatment of peptic ulcer is due to undergo a further revolution with
the discovery of Helicobacter pylorii as the causative organism. In a press
release following the VIIth Workshop on Gastroduodenal Pathology and
Helicobacter pylorii held in Texas in September 1994, the detailed results of a
trial by Dr. R.P.H. Logan of The Queen's Medical Centre, Nottingham, UK,
were presented in detail; they are given below:
Dr. Logan presented I-year follow-up data on a multicenter study of 154
patients with duodenal ulcers and H. pylorii infection. Six months data from
the same study were presented in May during Digestive Disease Week in New
Orleans.
Participants in the trial received either 500 mg clarithromycin (three times
a day) or placebo along with 40mg omeprazole (once a day) during the first 2
weeks of the study; all study participants received 40 mg omeprazole for an
additional 2 weeks.
According to Dr. Logan's 6-months interim results, ulcers healed after 4
weeks in 100% (64/64) of patients receiving clarithromycin and omeprazole
and 99% (71/72) of patients receiving omeprazole and placebo. However, a
significant difference was observed between the two study arms in the eradica-
tion of H. pylorii at 4-6 weeks after the conclusion of therapy. Patients who
were treated with the clarithromycin/omeprazole combination showed H.
pylorii rates of 83% - or 50 out of 60 evaluable patients - at 4-6 weeks post-
treatment versus 1 % (1174) in the group receiving placebo/omeprazole.
Furthermore, 6 months after treatment only 4% (2/53) of patients who
received clarithromycin and omeprazole experienced ulcer recurrence, versus
55% (36/60) of those who received placebo and omeprazole.
One year after treatment, the final results indicate incidence of ulcer
recurrence was 6% (3/51) in patients treated with clarithromycin and
omeprazole, versus 76% (47/62) of patients who received a placebo
and omeprazole. These data suggested that dual therapy with clarithromycin
and omeprazole might provide an effective regimen to heal and prevent occur-
rence of ulcers (LOGAN 1994).
The clarithromycin and omeprazole combination appeared to be well
tolerated according to researchers. Dr. Logan reported that only four patients
receiving clarithromycin and omeprazole and one patient receiving
omeprazole with placebo discontinued therapy because of adverse events.
Adverse events that occurred more frequently in the clarithromycinl
omeprazole-treated patients were tongue discoloration and taste perversion.
In this study, no patient experienced a serious adverse event.
At the Xth World Congress of Gastroenterology held in Los Angeles (2-
7 October 1994), a paper by GUSTAVSON et al. was presented. This dealt with
the pharmacokinetic interactions between omeprazole and clarithromycin in
20 male healthy volunteers. The results of their study led the authors to draw
three salient conclusions, namely:
242 J.P. GRIFFIN and P.F. D'ARCY
ing of daily blood glucose fluctuations. The antidiabetic effect does not involve
the stimulation of insulin secretion. Gastrointestinal problems are the most
commonly reported adverse effects in clinical trials. These include flatulence,
abdominal distension and diarrhoea. They are caused by fermentation of
unabsorbed carbohydrate in the bowel (CLISSOLD and EDWARDS 1988; NEW
MEDICINES 1994).
Currently, acarbose is licensed in the United Kingdom solely for the
treatment of non-insulin-dependent diabetes inadequately controlled by diet
alone or diet and oral hypoglycaemic agents. Although it has well-documented
antidiabetic properties, its precise place in therapy remains to be established.
It is not clear which patients should receive acarbose in place of, or in addition
to, established oral hypoglycaemic agents. It has been suggested that acarbose
may be useful for patients receiving an oral hypoglycaemic agent at the top of
the dosage range as an alternative strategy to commencing insulin. Further
clinical experience is required to establish whether it should be used in place
of traditional oral agents or whether it will act synergistically in combination
with them.
J. Cancer Chemotherapy
It has become standard practice to treat various malignant diseases with
cocktails of intensive combination regimens. For example in the treatment
of myeloma a combination of melphalan, cyclophosphamide, carmustine,
vincristine and prednisolone (M2) has been used. Other regimens combining
vincristine, doxorubicin and a corticosteroid, either dexamethasone (VAD) or
methylprednisolone (VAMP) have been developed.
In Ewing's sarcoma a protocol combining chemotherapy with cyclophos-
phamide, vincristine, dactinomycin and doxorubicin (VACA) with local radio-
therapy has achieved a 5-year survival rate. Other combination regimens have
been used for special tumours, e.g., CHAP 5, cisplatin, cyclophosphamide,
altretamine and doxorubicin combined with surgery for ovarian carcinoma.
Two of the better known cocktails are the MOPP regimen of mustine,
vincristine, procarbazine and prednisolone used in Hodgkin's disease, and the
COAP combination of cyclophosphamide, vincristine, ara-AC (fazarabine)
and prednisolone used in acute leukaemias.
Cocktails for use in malignant conditions have been developed by special-
ists in the field, partly on good therapeutic experience and partly empirically.
Where there is synergy, the mechanisms are often obscure. In many cases the
regimens have been developed from the philosophy of throwing into the
treatment everything in the pharmacopoeia that logically might work, and
avoiding two agents with the same mechanism of action on the malignancy.
From that initial experimental philosophy over the last 20 years there has
developed a rational approach to the evolution of the chemotherapeutic
cocktails.
246 J.P. GRIFFIN and P.F. D'ARCY
References
Alberti KGMM, Hockaday TDR (1987) Diabetes mellitus. In: Weatherall DJ,
Ledingham JGG, Warrell DA (eds) Oxford textbook of medicine vol 1, 2nd edn.
Oxford Medical, Oxford, pp 9.51-9.101
Synergistic Drug Interactions 247
Reynolds JEF (ed) (1993) Martindale, the extra pharmacopoeia. Pharmaceutical Press,
London, pp 137-138, 151-152
Ridgway E, Stewart K, Rai G, Kelsey MC, Bielawska C (1991) The pharmacokinetics
of cefuroxim axetil in the sick elderly patient. J Antimicrob Chemother 27:663-668
Rosenberg HA, Bates TR (1976) The influence of food on nitrofurantoin bio-
availability. Clin Pharmacol Ther 20:227-232
Sales JEL, Sutcliffe M, O'Grady F (1972) Cephalexin levels in human bile in presence
of biliary tract disease. Br Med J 3:441-443
Shanks RG (1979) Cardiac dysfunction. In: D'Arcy PF, Griffin JP (eds) Iatrogenic
diseases, 2nd edn, Oxford Medical, Oxford, pp 116-131
Sneader W (1985) Drug discovery: the evolution of modern medicines. Wiley,
Chichester
Soler NG, Pentecost BL, Bennett MA, Fitzgerald MG, Lamb P, Malins JM (1974)
Coronary care for myocardial infarction in diabetes. Lancet 1:457-477
Tsai SC, Burnakis TG (1993) Aldose reductase inhibitors: an update. Ann
Pharmacother 27:751-754
University Group Diabetes Program (1970) A study of the effects of hyperglycemic
agents on vascular complications in patients with adult onset diabetes. II.
Mortality results. Diabetes 19 [Suppl]:789--830
Van Gerven J, Lemkes H, van Dijk J (1992) Long-term effects of tolrestat in
symptomatic diabetic sensory polyneuropathy. J Diabetes Complications 6:45-48
Van Peer A, Westenborghs R, Heykants J, Gasparini R, Gauwenbergh G (1989) The
effect of food and dose on the oral systemic availability of itraconazole in healthy
subjects. Eur J Clin Pharmacol 36:423-426
Zenon G, Abobo C, Carter B, Ball D (1990) Potential use of aldose reductase
inhibitors to prevent diabetic complications. Clin Pharm 9:446--457
CHAPTER 9
In Vitro Drug Interactions
J.e. McELNAY and e.M. HUGHES
A. Introduction
In vitro drug interactions may be defined as those interactions which occur
outside the body. Drug interactions which will be discussed in this chapter are
those which occur between drugs due to reasons of incompatibility (e.g., drug-
drug interactions in an intravenous infusion), due to interaction of a drug with
its packaging (e.g., drug binding to an infusion bag), due to loss of drugs during
laboratory analyses (e.g., binding to laboratory equipment) or due to changes
in the bioavailability of drugs when the formulation is altered (McELNAY and
D'ARCY 1980).
B. Incompatibility Interactions
This form of interaction is often described as an incompatibility between two
agents and may be physical or chemical in nature. The interactions often occur
in solution, e.g., following addition of drugs to intravenous fluid containers,
after mixing drugs in syringes or in liquid preparations for oral or topical
administration. Physical incompatibility may be due to immiscibility or insolu-
bility. For example, addition of high concentrations of electrolytes to mixtures
in which the vehicle is a saturated aqueous solution of a volatile oil causes the
oil to separate and collect as a surface layer. This is the case with potassium
citrate mixture BPC, in which the large quantity of soluble solids salts out the
lemon oil; quillaia tincture is therefore added as a suspending and emulsifying
agent to prevent this (CARTER 1975).
Chemical incompatibility may be caused by pH change, decomposition or
complex formation. A prime example of this type of interaction involves the
barbiturates, e.g., pentobarbitone and phenobarbitone. Solutions of the salts
of these compounds are very alkaline and incompatible with acids, acidic salts
and acidic syrups, e.g., lemon syrup, which may precipitate the corresponding
insoluble barbituric acid derivative (CARTER 1975). The BRITISH PHARMACEUTI-
CAL CODEX (LUND 1994) documents a number of physical and chemical inter-
actions between drugs and other medicinal products; these are summarised in
Table l.
Many such incompatibilities are no longer viewed as being particularly
problematic as far as oral formulations are concerned since easy solutions to
250 J.e. McELNAY and e.M. HUGHES
the problems can usually be found. There are often, however, many contem-
porary reports in the pharmaceutical literature on drug interactions due to
incompatibility in intravenous fluids. This can either be due to an interaction
between two or more drugs added to intravenous fluids prior to administration
to patients, e.g., in the case of cancer chemotherapy or due to a single added
drug being incompatible with the intravenous vehicle. GRIFFIN and D' ARCY
(1988) have prepared a list of such incompatibilities, for example, ampicillin
should not be mixed with amino acids [constituents of total parental nutrition
(TPN) regimens] as immunogenic and allergic conjugates could be formed;
amphotericin is incompatible with all electrolytes and, thus, must be reconsti-
tuted with water and infused with 5% glucose (GRIFFIN and D'ARCY 1988).
ALLWOOD (1994) described the problem of drug-drug co-precipitation, which
may be particularly problematic in intravenous therapy. Such interactions
In Vitro Drug Interactions 251
arise due to the mixing of organic anions and cations, which form ion pairs.
Examples of drugs at risk of forming ion pairs with other drugs include
gentamicin and other aminoglycosides, with heparin and some cephalosporins.
One of the areas of medicine where simultaneous co-administration of a
number of intravenous medicines is a clinical necessity is cancer chemo-
therapy. FOURNIER et al. (1992) studied the effect of a commercial formulation
of 5-fluorouracil on the stability of cisplatin and carboplatin to determine
whether the drugs could be mixed in containers or intravenous lines. When
cisplatin was incubated with 5-fluorouracil, high-performance liquid chroma-
tography (HPLC) studies demonstrated a rapid disappearance of the parent
platinum compound, the extent of degradation being 75% after 3.5h. It was
found that the degradation was in fact caused by an interaction between
cisplatin and trometamol, the excipient used in the 5-fluorouracil formulations
in some countries to buffer the solution at pH 8.2. This interaction resulted in
complete inhibition of the antitumour activity of cisplatin in mice. When
cisplatin was incubated at the same pH in trometamol-free sodium hydroxide
solutions, the parent compound was transformed into an active compound that
was toxic to mice. Degradation of carboplatin-trometamol admixtures was
similar to that found for cisplatin, but occurred at a slower rate. As in the case
of cisplatin, incubation of carboplatin in a sodium hydroxide solution resulted
in toxic effects. It was concluded that both carboplatin and cisplatin were
incompatible with 5-fluorouracil formulations containing trometamol.
Due to the possibility of incompatibility with constituents, as a general
rule no drugs should be added to TPN fluids. To avoid direct contact between
TPN fluids and drugs administered by the intravenous route, COLLINS and
LUTZ (1991) have examined the co-administration of phenytoin and TPN
mixtures in multilumen catheters. These devices can be used to reduce the
need for frequent venipuncture and provide a method of administering incom-
patible drugs. The study utilised an in vitro model flow system to examine the
physicochemical phenomena that occurred when these two incompatible enti-
ties (phenytoin and TPN fluid) were simultaneously administered through
multilumen catheter systems into a circulating fluid (sodium chloride, sodium
bicarbonate and albumin in solution). Double- and triple-lumen catheters
were used as shown in Fig. 1. Video recordings were made of the drug flow and
assays of phenytoin concentration were performed on samples of the circulat-
ing fluid. White clouds of phenytoin precipitation were seen near the tip of the
double-lumen catheter, but not the triple-lumen device. Infusion through the
former resulted in an average of 6% loss of phenytoin which, on microscopic
examination, appeared as spindle-shaped crystals. In some cases, millimetre-
size fragments of phenytoin precipitate were seen to dislodge from the tip of
the double-lumen catheter. It was suggested that the interaction was due to the
close proximity of the two orifices at the end hole of the double-lumen cath-
eter, which permitted mixing of the two effusing streams of the incompatible
agents. The staggered orifices of the triple-lumen catheter reduced this inter-
action greatly; no crystal fragments were observed, although a thin coating of
252 J.C. McELNAY and C.M. HUGHES
A.
Double
lumen
Call'leter
B
Tnpl~ Lumen
Catheter
Fig. 1. Drawings of tip of two types of catheters used in study of concomitant admin-
istration of phenytoin and TPN fluid. (After COLLINS and LUTZ 1991)
white film did form near the opening of the proximal and middle side holes of
the catheter. Although the authors recognised the value of these multilumen
devices, they did advise caution in their use, particularly in the clinical setting,
with combinations of incompatible drugs. To be forewarned is to be forearmed
and indeed the best approach to prevention of drug-drug incompatibility
interactions is to use published information on the physical and chemical
properties of medicinal agents which are to be used concomitantly and avoid
their coming into direct contact.
N
Ul
Ul
N
Ul
0\
Table 3. Continued
Chlopromazine Sorbed to PVC Use alternative form of plastic for AIRAUDO et al.
Clomipramine Sorbed to PVC administration, e.g., Stedim infusion bags (1993)
Chlorazepate dipotassium Sorbed to PVC with polyethylene lining
salt
Vitamins
Vitamin A Sorbed to PVC Use vitamin A palmitate which CHIOU and
does not sorb to PVC MOORHATCH
(1973)
Vitamin E Sorbed to plastic materials in Use glass DROIT et al. (1991)
infusion sets (material not
specified)
Vitamin D (ca1citriol) Sorbed to PVC Increase amount of surfactant used in LI et al. (1992)
formulation as this reduces amount of free
drug available for sorption. Avoid PVC ......
Miscellaneous agents (1
Miconazole Sorbed to PVC Use glass; if PVC must be used, drug must HOLMES and
be prepared and administered immediately ALDOUS (1991) ~
t!j
Bupivacaine Sorbed to glass and plastic Avoid alkalinisation as sorption effects BOMHOMME et al. t"'
1. Sorption
Sorption is probably the most important underlying mechanism responsible
for a number of drug interactions associated with pharmaceutical packaging.
This term describes loss of drug to the interacting material and includes
adsorption to the surface plus absorption of the drug into the body of the
material. Adsorption is characterised by rapid and substantial initial drug loss
associated with binding to the surface, leading to surface saturation. Adsorp-
tion can occur to all solid surfaces irrespective of the nature of the material
(ALLwooD 1994). Absorption is defined as the migration of the drug into the
matrix of the interacting material (usually plastic) and an equilibrium becomes
established between liquid and solid phases. The process develops more slowly
than adsorption, but saturation occurs eventually, although it may take many
hours (ALLwooD 1994). The plastic that is most often implicated in such
interactions is polyvinyl chloride (PVC), although other materials can also be
involved.
When examining sorption interactions, the physicochemical properties of
the drug in question must be considered: the extent of ionisation and lipid
solubility have been proposed as two determining factors of sorption by plastic
infusion bags (KOWALUK et al. 1981). A number of models have been devel-
oped to predict solute sorption by PVc. ROBERTS et al. (1991) estimated the
time course of the sorption of drugs using a diffusional model in which the
plastic was assumed to act as an infinite sink. This model appeared to be
suitable for estimation of storage times relevant to clinical usage and enabled
the magnitude of the uptake in a specified time to be described by a single
parameter known as the sorption number. This sorption number was defined
by the plastic-infusion solution partition coefficient, the diffusion coefficient in
the plastic, the fraction of drug unionised in solution, the volume of the
infusion solution and the exposed surface area of the plastic. This parameter
(sorption number) could be used to predict the effects of a number of factors,
e.g., time, plastic surface area, solution volume and solution pH on fractional
drug loss. JENKE (1993) developed a similar model in which storage time
proved to be a critical factor in predicting sorption; at longer storage times, it
was found that the infinite sink approach as described by ROBERTS et al. (1991)
was inaccurate, while at shorter storage times the sorption profile for a solute
can be estimated from its hexane-water and octanol-water partition coeffi-
cients via a simple diffusion-based expression. It was noted that differences
between observed and predicted behaviour illustrated the problems in a theo-
retical approach; thus, it is only in the experimental or clinical environment
that accurate assessment relating to drug sorption can be made.
Not all sorption interactions will be clinically significant. The schematic
representation of an arbitrary time scale (Fig. 2) illustrates that those inter-
258 J.e. McELNAY and C.M. HUGHES
•+
Nitroglycerin Vitamin A Benzodiazepine Lignocaine
Itt Chlormethiazole
t
24 48
Insulin Diazepam Warfarin
Fig. 2. Time-scale for clinically significant losses of drug due to sorption to plastics
materials (ALLWOOD 1995, personal communication)
actions occurring close to time 0 are clinically significant, whereas those occur-
ring at the other end of the scale may be considered insignificant. Insulin and
the nitrates are examples of drugs which are sorbed to a clinically significant
degree; interactions involving these drugs are discussed below.
a) Insulin
The non-specific surface binding of insulin from dilute solutions was first
reported by FERREBEE et al. (1951), who showed that the polypeptide was
strongly adsorbed by laboratory glassware. Since then, further work has shown
that insulin also binds to siliconised glassware, paper and polypropylene
(NEWERLY and BERSON 1957; HILL 1959; PETIY and CUNNINGHAM 1974).
WIDEROE et al. (1983) evaluated the degree of insulin adsorption to plastic
containers during continuous ambulatory peritoneal dialysis (CAPD). Ap-
proximately 65 % of insulin which was added to the dialysis fluid was sorbed to
the plastic container.
A series of studies by TWARDOWSKI et al. (1983a-c) examined this sorption
phenomenon in greater depth. Binding of insulin to the plastic of dialysis
solution containers was instantaneous and apparently not influenced by time
of contact. Further experimental work revealed that insulin binding could be
partly reversed by simple washing with water (TWARDOWSKI et al. 1983b). A
third study demonstrated that insulin sorption was influenced by temperature:
more insulin was bound at 37°C than at 24°C (TWARDOWSKI et al. 1983c).
McELNAY et al. (1987) studied the binding of human insulin to three types
of burette intravenous administration sets - a Standard set (cellulose propi-
onate burette and PVC tubing), an Amberset (cellulose propionate burette
and PVC tubing) and a Sure set (methacrylate butadiene styrene burette and
polybutadiene tubing) over a 6-h period. The latter administration set had
been previously shown to resist sorption of a number of drugs (LEE 1986).
However, in this case, the Sureset bound insulin more extensively than the
Standard set or Amberset as illustrated in Fig. 3. It was suggested that, in the
clinical setting, this adsorption would present a particular problem since dur-
ing intravenous infusion the concentration of drug is relatively low and the
surface area available for binding is relatively high. This non-specific adsorp-
tion of insulin can best be approached by administering insulin in a small
volume via a syringe pump as the surface area is much reduced compared with
the total amount of insulin present or, alternatively, more drug than is re-
quired is added to the infusion container and blood sugar levels closely moni-
In Vitro Drug Interactions 259
25
20
'"
E
0 15
0
-- 10
I/)
::J
~ 5
0
o 0.083 0.5 2 4 6
a Time (Hours)
c: 25
-cE
.2
..,-
.. '" 20
Gl o 15
go
01/)
0--
10
c: =!
3;':; 5
.='" 0 ~ ~ r1r.;:l
~
o 0.083 0.5 2 4 6
b Time (Hours)
Fig. 3. a Concentration versus time profile for human insulin when stored in burette
chambers of a standard set (0), Amberset (~) and Sureset (~ burette administration
sets. • represents mean control data at zero time. (Maximum coefficient of variation
within replicate data points was 3.2%.) b Concentration (± SD) versus time profile for
human insulin when stored in administration tubing from a standard set (0), Amberset
(~) and Sure set (~) burette administration sets. • represents mean control data at
zero time. (After McELNAY et al. 1987)
tored. It has also been suggested that human serum albumin or hydrolysed
gelatin (polygeline) can be used as a carrier for insulin, since the binding to
these agents exceeds that of the binding to surfaces (SCHILDT et al. 1978). This
approach is, however, not used routinely.
b) Nitrates
A number of studies have reported interaction of a range of nitrates, e.g.,
glyceryl trinitrate and isosorbide dinitrate with packaging and intravenous
delivery equpiment. A number of mechanisms of interaction appear to take
place simultaneously with the nitrates including adsorption, absorption and
permeation.
used to examine the loss of drug efficacy. It was concluded that glyceryl
trinitrate was removed from aqueous solution by plastic via an absorptive
process, suggesting a degree of penetration into the plastic. It is also likely, due
to the volatile nature of glyceryl trinitrate, that it penetrates through the
plastic and is lost by evaporation from the plastic air interface.
The loss of glyceryl trinitrate was confirmed by BAASKE et al. (1980), who
studied the effect of intravenous filters, containers and administration sets on
glyceryl trinitrate efficacy. Filters decreased glyceryl trinitrate concentrations
by 2%-55%, whereas storage in glass bottles over 48h had no effect. Storage
in plastic intravenous infusion bags led to substantial concentration decreases
that were related to surface contact area and temperature. Glyceryl trinitrate
solution was also infused through an administration set, resulting in an imme-
diate and substantial reduction in drug concentration. Using a theoretical
approach, MALICK et al. (1981) described a model of glyceryl trinitrate loss in
which the drug is adsorbed onto the material surface and then partitioned into
the plastic.
Work has continued on the glyceryl trinitrate sorptive phenomenon.
LOUCAS et al. (1990), for example, examined the sorptive properties of glyceryl
trinitrate in relation to PVC, with particular reference to the role of the
admixture vehicle. Initially (first 10min of sampling) the loss of glyceryl
trinitrate to the PVC administration set was greatest from dextrose admix-
tures, intermediate for water admixtures and least for saline admixtures. Be-
tween 15 and 20 min of the experimental period, the greatest loss of glyceryl
trinitrate was noted for saline. It was concluded that glyceryl trinitrate avail-
ability in admixtures in contact with PVC was dependent on the ionic strength
of the vehicle and the time at which measurements were made.
The loss of glyceryl trinitrate during passage through a PVC or a polyeth-
ylene infusion set has also been investigated in relation to concentration and
flow rate (HANSEN and SPILLUM 1991). The concentration of glyceryl trinitrate
had no influence on the loss of drug, although the greatest loss occurred at the
slowest rate of infusion. A loss of 70% of glyceryl trinitrate during infusion
using PVC apparatus was found, whereas the loss when using the polyethylene
set did not exceed 15% over 8h.
As PVC appears totally unsuitable as a material in which to store glyceryl
trinitrate, other forms of plastics have been used. SALOMIES et al. (1994)
measured the sorption of glyceryl trinitrate, diazepam and warfarin sodium (in
normal saline) by a new polypropylene-lined infusion bag (Softbag) and com-
pared this to sorption in glass bottles and PVC bags. The containers were
stored at room temperature without protection from light for 24h (diazepam)
and 120h (glyceryl trinitrate and warfarin). The three drugs did not demon-
strate any sorption to glass bottles or the Softbag containers although sorption
was high to PVC (Fig. 4). The polypropylene-lined Softbag therefore provides
a useful approach in the avoidance of sorption of these agents.
ALTAVELA et al. (1994) viewed that loss of glyceryl trinitrate onto admin-
istration sets may have little impact on the care of patients as infusions of the
In Vitro Drug Interactions 261
100
80
roo-----------I
e,
riia
.".,,~
o
0.
o
'"
.... 60
o
~ 40
20
o {L_ _ _ _- L____- L____- L____- L____ __ ~ ~
o 5 10 15 20 25
a Time (hours)
100 ift3:..--~-g-~--O-g
c
.;: 80 ~-e
'"
U
\
e
""
>.
60
-
CI
~
z 40 e'-...........
e_e_
~ e-e
20
o 20 40 60 80 100 120
b Time (hours)
--"
E T"
~
"0
0
80 e - T _ " ___ T
en \
e
20
o{
0 20 40 60 80 100 120
C Ti m e (h 0 u rs)
drug are adjusted for the patient's response and thus drug loss may be of little
significance. Another consideration was the expense of polyethylene sets com-
pared to PVC sets; thus this group randomly assigned patients with ischaemic
heart disease to receive glyceryl trinitrate solution through either a polyethyl-
ene administration set or a PVC set. It was found that patients who received
glyceryl trinitrate through the PVC set had the same clinical response as
patients who had received the drug through the polyethylene set and there
were no differences between the initial dosage adjustment time, the number of
dosage adjustments and indications for them, the duration of the infusion, the
time of tubing changes and total dose of glyceryl trinitrate.
d) Isosorbide Dinitrate
This long-acting nitrate has been used orally, sublingually and intravenously
in the treatment and prevention of angina and in congestive heart failure
therapy. As stated in its current United Kingdom data sheet, isosorbide
dinitrate is incompatible with PVC containers, but may be used with glass and
polyethylene (ASSOCIATION OF BRITISH PHARMACEUTICAL INDUSTRY 1994).
COS SUM and ROBERTS (1981) investigated the availability of this drug from
intravenous delivery systems. Appreciable loss of the drug was found follow-
ing storage in PVC infusion bags and/or in burettes (cellulose propionate) of
administration sets. This degree of loss was also related to the flow rate of the
infusion; lower rates of recovery of the nitrate were found at slower flow rates.
No loss of isosorbide dinitrate was found during the storage of the drug in glass
for 200h.
SAUTOU et al. (1994) evaluated the compatibility of isosorbide dinitrate
and heparin with polypropylene, polyethylene and PVC under simulated clini-
cal conditions. Results indicated that both drugs were compatible with
polypropylene syringes; preparing the syringes 8h in advance did not alter the
stability of the drugs, provided that the nitrate was not refrigerated. The
concentration of isosorbide dinitrate infused through PVC/polyethylene tub-
ing (PVC on the outside and polyethylene on the inside) was unaffected,
irrespective of whether it was administered alone or together with heparin. As
was expected, both drugs were sorbed by the PVc. The nitrate was rapidly
fixed to the PVC and then released again; this release occurred earlier when
heparin was co-administered. It was concluded by the authors that adsorption
and release of the drugs appeared to be very random.
2. Leaching
The term leaching is applied to the migration of a substance from packaging
material into a medicine. It is influenced in rate and extent by the solvent
system used in the preparation, and pH and temperature conditions during
processing and storage (COOPER 1974). Examples are the leaching of alkali
from soda-glass bottles and the leaching of zinc salts, used as activators, from
rubber closures. Barium ions leached from borosilicate glass may react with
sulphates of drugs such as kanamycin, atropine or magnesium to form barium
In Vitro Drug Interactions 263
3. Permeation
Permeation is another physicochemical mechanism of drug binding associated
with some forms of pharmaceutical packaging. It is a combination of absorp-
tion followed by migration to the outer surface, from which the drug is
released by evaporation. Losses may be substantial and they continue
throughout the period of administration since saturation of the plastic is never
achieved (ALLWOOD 1994). Permeation may also occur due to passage of
gases, vapours and liquids through packaging; water vapour permeates
through silicone rubber, and to varying degrees through plastics, particularly
PVC and polystyrene (LUND 1994).
In a study conducted by KOWALUK et al. (1981) a number of hypnotic
agents were investigated in relation to incompatibility with PVC infusion bags.
Chlormethiazole and diazepam were found to be substantially lost after 24h of
storage (33% and 20%, respectively). The chlormethiazole was shown to
penetrate through the plastic as evidenced by a distinctive odour of the drug in
the immediate vicinity of the bag. COS SUM and ROBERTS (1981) also reported
a loss in drug concentration when chlormethiazole was stored and infused
through PVC bags and infusion sets; the average loss from the infusion fluid
approximated to 15%-30%. Loss was negligible when infused from glass
syringes through polyethylene tubing, thereby presenting an alternative to
PVC materials. As mentioned above the loss of glyceryl trinitrate from intra-
venous fluid containers involves a degree of permeation through plastic mate-
rials followed by loss by evaporation (particularly in the case of PVC). This
phenomenon is not limited to intravenous preparations of the drug, i.e., sublin-
gual glyceryl trinitrate should be stored in glass bottles.
4. Polymer Modification
In the work of KOWALUK et al. (1981) described in the previous section, it was
also reported that the PVC infusion bags containing chlormethiazole became
In Vitro Drug Interactions 265
I. Cyclosporin
This cyclic peptide is widely used in organ and tissue transplantation and is
associated with a number of side effects, most notably nephrotoxicity (CALNE
266 J.e. McELNAY and e.M. HUGHES
II. Chloroquine
GEARY et al. (1983) showed that in the laboratory setting preparations of
chloroquine in various solutions showed decreases in concentration of up to
40% when stored in glass containers and that the drug was extensively bound
to cellulose acetate filters. YAHYA et al. (1985) examined the binding of
chloroquine to different grades of laboratory glassware at different concentra-
tions and different pH conditions. Storage in soda glass (test tubes and glass
wool) showed a decrease in original drug concentration of up to 60% and 97%,
respectively. The highest degree of binding was recorded at physiological
pH (7.4) and at low concentrations. Borosilicate glass did not exhibit any
chloroquine binding. A second study investigated the binding of this anti-
malarial agent to a range of plastic materials under conditions of varying pH
and concentration (YAHYA et al. 1986). Chloroquine was sorbed by cellulose
propionate, ethylvinyl acetate, methacrylate butadiene styrene, polyethylene,
polypropylene and PVC, but not by high-density polystyrene. Passing
chloroquine solutions through cellulose acetate filters once resulted in a high
loss of drug. The unionised form of the drug was preferentially sorbed. These
studies indicate that care must be taken when quantifying chloroquine in the
laboratory since the drug will come into contact with a range of materials. This
is particularly the case in kinetic studies and malarial sensitivity testing, when
chloroquine may be bound to plastic syringes or pipettes during sampling;
binding to membrane filters used to clarify and/or to sterilise chloroquine
solutions prior to use or analysis could also give rise to erroneous results
(YAHYA et al. 1986). Although this problem has been highlighted for
chloroquine, it is well known that care must be taken in the manipulation of
other drugs in the laboratory setting, e.g., some benzodiazepines are highly
bound to glassware and this necessitates the silylation of glassware prior to
use.
this may result in changes in bioavailability of the active drug constituent and
there have been a number of reported cases where this has led to adverse
clinical consequences for patients (McELNAY and D' ARCY 1980). Other re-
ports are from experimental studies which stress the importance of formula-
tion in relation to bioavailability.
The two classical examples of this type of in vitro effect involve the drugs
phenytoin and digoxin. Anticonvulsant intoxication resulted from subtle
changes to the formulation of phenytoin capsules (TYRER et al. 1970); lactose
replaced calcium sulphate as an excipient, resulting in increased dissolution
and higher serum levels of phenytoin as illustrated in Fig. 5. Particle size
changes to digoxin also led to marked changes in digoxin levels (JOHNSON et al.
1973). Tablets manufactured after May 1972 contained a smaller particle size
digoxin, which resulted in a higher degree of absorption of the drug and
overdigitilisation of patients. This led to much stricter statutory controls in
relation to formulation in developed countries. However, a later study by
FLETCHER and SUMMERS (1980) revealed that these controls did not extend to
all countries. Five brands of digoxin, which were available in South Africa at
the time, were compared in relation to physical and dissolution characteristics
and bioavailability. Only one brand satisfied all requirements, based on official
specifications. One brand did not satisfy diameter specifications, was soft and
crumbled easily, and showed only 34.82% dissolution within the period exam-
ined. A second brand was viewed as a very low grade product which neither
dispersed readily nor complied with the specifications for digoxin content. It is
this type of report which has given rise to concerns over the quality of some
generic products available in developing countries. For most products, this is
~ ~
30 "T1
-:25
.....
I
--
E -0
i"
[ 20
c:
~:i"
o.§ 15 30 CD
>-
c: ~
II iii
20 5-
...
~IO
::J
8 5 10
'3
<= «&
0 0
!
8 16 2'1 32 o 8
Doy
Fig. 5. Blood phenytoin concentrations in a patient taking phenytoin (400mglday),
with excipients respectively as shown (lactose, calcium sulphate, lactose). Vertical
columns represent daily faecal excretion of phenytoin when measured. (After TYRER
et al. 1970)
268 J.e. McELNAY and C.M. HUGHES
1.0
.9
.8
.7
.6
.5
Theophylline Solullon
_-_TheophyISR N~14
.2 cr - ~ Aerolate N ~4
.---.. Theobld N~6
.1
O~-L~~-L--~~~~~~~~~~~--
o 4 6 8 10 12 14 16 18 20
a Time (hours)
::~ :r-~==I
I
1.0 ................... T .. __ ...
,-.---- ----. 1
.9 ".1_+-.1':::-1-'-1
.8
1''' ,,1--1
I .
..
1)
III
.Q .7 /Y'{
0
~),/
ll·
CI)
.Q .6
0:(
'-e-
c: .5
0
( ,)
.4
t;f. Theophylline Solullon
r~
4-
.3 SUSTAINED RELEASE TABLETS
. - ... Theodur-l00mg/ N~5
.2 t{ 0 - - ~ Theodur· 300mg I
..---. Aminodur N::5
.1
0
2 4 6 8 10 12 14 16 18 20 22 24 26 28
b Time (hours)
Fig. 6. a Absorption of sustained-release bead-filled capsules of theophylline. b Ab-
sorption of sustained-release tablets of theophylline. Aminodur, Aerolate and Theobid
are seen to have suboptimal absorption. (After WEINBERGER et al. 1978)
270 J.e. McELNAY and e.M. HUGHES
portions of PVA and PVA-MA it was possible to affect the release character-
istics of the drug. Tablets made with crystalline PVA provided instant release
of theophylline in vitro, whereas those made with a larger proportion of PVA-
MA relative to PVA demonstrated a very prolonged release profile. It was
concluded that the crystallinity of the polymer played a major role in the
release of the drug, as high-crystallinity polymers (PVA) are less permeable to
diffusion than low-crystallinity polymers (PVA-MA); the latter are known to
dissolve more slowly in water and hence drug release is sustained. The
bioavailability (in dogs) of a formulation containing PVA:PVA-
MA: theophylline in the ratio of 1: 9: 10 was equivalent to that obtained after
administration of a commercially available product Theo-Dur.
As previously stated, theophylline is a drug which is normally prescribed
by brand name to ensure that the patient receives the same release/absorption
patterns of the drug each time it is administered. A recent study by HENDELES
et al. (1995) evaluated the relative bioavailability and clinical efficacy of a
generic slow-release theophylline tablet and Theo-Dur in 14 adults with
asthma. The results of the study indicated that the generic slow-release theo-
phylline tablet and Theo-Dur were not bioequivalent. Although they were
absorbed to the same extent, as indicated by similar areas under the serum
concentration-time curves and peak concentrations, the generic product was
absorbed more rapidly. This was demonstrated by the shorter time to peak
concentration, larger fluctuations between peak and trough concentrations
and slightly lower mean trough concentration. Therapeutic equivalence was
also assessed using attenuation of the response to exercise as a surrogate for
clinical effect (exercise challenge was 17 h after last dose of theophylline).
Although neither product effectively attenuated exercise-induced
bronchospasm, the authors considered that despite the differences in absorp-
tion rates both formulations were therapeutically equivalent, but not
bioequivalent.
As a result of stricter statutory controls with regard to changes in product
formulation, there are now relatively few clinical reports of drug interactions
arising from formulation modification.
excipient povidone may complex with anionic or cationic dyes, and drugs such
as chlorpromazine and chloramphenicol. Gels made with povidone may in-
crease in viscosity as a result of complexation with 8-hydroxyquinolone
sulphate or thiomersal sodium. Excipients (or another drug) may also affect
the stability of a drug, leading to reduced bioavailability and changed thera-
peutic efficacy as reported by the BRITISH PHARMACEUTICAL CODEX (LUND
1994): sodium metabisulphite rapidly inactivates cisplatin and propylene gly-
col and macro go Is catalyse the degradation of benzoyl peroxide to benzoic
acid and carbon dioxide.
An early study by MCGINITY and LACH (1976), using dissolution and
dialysis techniques, examined the adsorption of various drugs to montmorillo-
nite clay; this colloidal magnesium aluminium silicate clay has been used as a
disintegrant, binder and lubricant. Chlorpheneramine maleate, amphetamine
sulphate and propoxyphene hydrochloride were all found to be strongly
bound to montmorillonite due to their cationic nature; amphoteric compounds
such as theophylline and caffeine were found to bind moderately to the clay,
whereas sodium salicylate and sulphanilamide (anionic in nature) were re-
leased rapidly from montmorillonite, i.e., binding was minimal.
Although these latter binding interactions are largely regarded as detri-
mental in that drug absorption will be reduced, the binding can be used to
advantage in the formulation of sustained-release products. The interaction of
piroxicam, for example, with various synthetic polymers (polyethylene glycol
400,600,1000,4000,6000 and 20000, polyvinylpyrrolidone 30000, 40000 and
64000, and dextran 40000 and 75000) has been determined by ELSHAITAWY et
al. (1994). The binding of piroxicam in different concentrations with the differ-
ent polymers showed that increasing drug concentration was accompanied by
an increase in the amount of bound drug to the polymer, until near saturation.
The binding capacity of piroxicam was found to be greatest for the poly-
vinylpyrrolidones and least for dextran.
CHOWHAN and CHI (1986a) compared the two lubricants magnesium stear-
ate and sodium stearyl fumarate under identical conditions, in order to study
their roles in drug-excipient interactions; the drug in question was ketorolac.
After prolonged mixing, sodium stearyl fumarate did not affect the drug of
excipient (crospovidone) and, as a result, the disintegration time and drug
dissolution from hand-filled capsules were not adversely affected. In contrast,
magnesium stearate did demonstrate a drug-crospovidone interaction which
resulted in an increased disintegration time and dissolution time. This resulted
from particle-particle interaction whereby lamination and flaking of magne-
sium stearate occurred. These flakes adhered to drug particles and caused a
significant reduction in the dissolution rate. In a further study, using
prednisone, the effect of powder mixing on drug-excipient interactions and
their effect on in vitro dissolution from capsules was studied (CHOWHAN and
CHI 1986b). Two powder formulations contained dibasic calcium phosphate
dihydrate as a filler and potato starch or sodium starch glycolate as a
disintegrant were studied. The third powder formulation contained
272 J.e. McELNAY and e.M. HUGHES
100
90
80
"0 70
OJ
>
~ 60
.!!l
"0
Ol 50
"
:J
'0 40
OJ
g> 30
C
OJ
(.)
Q; 20
11.
10
100
90
80
"0 70
OJ
>
(560
l:l
:0
Ol
50
2
~o 40
OJ
g> 30
C
OJ
(.)
F. Conclusions
Interpretation of in vitro drug interactions involves a number of basic prin-
ciples (ALLwooD 1994):
1. An understanding of the chemistry of drugs in the salt and/or free form and
the effects of pH
2. A knowledge of the basis for physicochemical interactions between solutes
and solids
3. Implementing practical solutions to overcome an in vitro drug interaction
4. Evaluation of the clinical effect of the in vitro drug interactions
Many of the interactions which have been discussed in this chapter may be
explained in chemical and physical terms. Once the interaction has been
elucidated a number of approaches can be used to overcome or avoid un-
wanted sequelae. Ways of overcoming the interactions include:
1. Avoidance of direct contact of interacting agents, e.g., via use of multiuse
cannulae for intravenous administration
2. Adding increased doses of drugs to infusion fluids when the drug is bound
to the infusion apparatus
3. Ensuring that oral formulations are adequately tested for bioavailability
prior to clinical use
4. Use of glass or suitable plastics which do not interact with drugs in question,
e.g. use of polypropylene-lined infusion bags and administration sets for
drugs which are highly bound to PVC
References
Airaudo CB, Gaytesorbier A, Bianchi C, Verdier M (1993) Interactions between six
psychotherapeutic drugs and containers: influence of plastic material and infusion
solutions. Int J Clin Pharmacol Ther 31:261-266
In Vitro Drug Interactions 275
Allwood MC (1986) The release of phthalate ester plasticizer from intravenous admin-
istrations sets into fat emulsion. Int J Pharm 29:233-236
Allwood MC (1994) Drug stability and intravenous administration, 2nd edn. Clinical
pharmacy practice guide. UKCPA, University of Leeds Media Service, Leeds
Altavela JL, Haas CE, Nowak DR, Powers J, Gacioch GM (1994) Clinical response to
intravenous nitroglycerin infused through polyethylene or polyvinyl chloride tub-
ing. Am J Hosp Pharm 51:490-494
Association of British Pharmaceutical Industry (ABPI) (1994) Data sheet compen-
dium 1994/95. Datapharm, London
Autain J (1963) Plastics in pharmaceutical practice and related fields, part I. J Pharm
Sci 52:1-23
Baaske DM, Amann AH, Wagenknecht DM, Mooers, M, Carter JE, Hoyt HJ, Stoll
RG (1980) Nitroglycerin compatibility with intravenous fluid filters, containers,
and administration sets. Am J Hosp Pharm 37:201-205
Benvenuto JA, Anderson RW, Kerkof K, Smith RG, Loo TL (1981) Stability and
compatibility of antitumor agents in glass and plastic containers. Am J Hosp
Pharm 38:1914-1918
Birely A W, Scott MJ (1982) Plastic materials, properties and applications. Blackie,
Glasgow, pp 1-18
Bonhomme L, Benhamou D, Beugre T (1992) Slow-release effect of pH-adjusted
bupivacaine: in vitro demonstration. Int J Pharm 84:33-37
British National Formulary (1994) No 28. British Medical Association and Pharmaceu-
tical Press, London
British National Formulary (1995) No 29. British Medical Association and Pharmaceu-
tical Press, London
Brydson JA (1982) Plastics materials. Butterworth, London, pp 18-143
CaIne RY, White DRG, Thiru S (1978) Cyclosporin A in patients receiving renal
allografts from cadaver donors. Lancet 11:515-524
Carter SJ (1975) Cooper and Gunn's dispensing for pharmaceutical students, 12th edn.
Pitman, London, pp 253-264
Chen ML, Chiou WL (1982) Adsorption of methotrexate onto glassware and syringes.
J Pharm Sci 71:129-131
Chiou WL, Moorhatch P (1973) Interaction between vitamin A and plastic intravenous
bags. JAMA 223:328
Chowhan ZT, Chi LH (1986a) Drug-excipient interactions resulting from powder
mixing. IV. Role of lubricants and their effect on in vitro dissolution. J Pharm Sci
75:542-545
Chowhan ZT, Chi LH (1986b) Drug-excipient interactions resulting from powder
mixing. III. Solid state properties and their effect on drug dissolution. J Pharm Sci
75:534-541
Collins JL, Lutz RJ (1991) In vitro study of simultaneous infusion of incompatible
drugs in multilumen catheters. Heart Lung 20:271-277
Cooper J (1974) Plastic containers for pharmaceuticals testing and control. WHO,
Geneva, pp 1-13
Cossum PA, Roberts MS (1981) Availability of isosorbide dinitrate, diazepam and
chlormethiazole from i.v. delivery systems. Eur J Clin PharmacoI19:181-185
DiLuccio RC, Hussain MA, Coffin-Beach D, Torosian G, Shefter E, Hurwitz AR
(1994) Sustained-release oral delivery of theophylline by use of polyvinyl alcohol
and polyvinyl alcohol-methyl acrylate polymers. J Pharm Sci 83:104-106
Drott P, Meurling S, Meurling L (1991) Clinical adsorption and photodegradation of
the fat-soluble vitamin A and vitamin E. Clin Nutr 10:348-351
Durig T, Fassihi AR (1993) Identification of stabilizing and destabilizing effects of
excipient-drug interactions in solid dosage form design. Int J Pharm 97:161-170
Elshattawy HH, Ahmed EI, Bayomi MA, Abdelfatah 1M (1994) Piroxicam synthetic
polymers interactions. Pharm Ind 56:396--399
Ferrebee JW, Johnson BB, Mithoefer JC, Gardella JW (1951) Insulin and adrenocor-
ticotropin labelled with radio-iodine. Endocrinology 48:277-283
276 J.e. McELNAY and e.M. HUGHES
Wideroe TE, Smeby LC, Berg KL, Jorstad S, Svartas TM (1983) Intraperitoneal (1 251)
insulin absorption during intermittent and continuous peritoneal dialysis. Kidney
Int 23:22-28
Yahya AM, McElnay JC, D'Arcy PF (1985) Binding of chloroquine to glass. Int J
Pharm 25:217-223
Yahya AM, McElnay JC, D'Arcy PF (1986) Investigation of chloroquine binding to
plastic materials. Int J Pharm 34:137-143
Yuen PH, Denman SL, Sokoloski TD, Burkman AM (1979) Loss of nitroglycerin from
aqueous solution into plastic intravenous delivery systems. J Pharm Sci 68:1163-
1166
CHAPTER 10
Age and Genetic Factors in Drug Interactions
J.e. McELNAY and P.F. D'ARCY
A. Introduction
It has long been known that specific patient factors can influence the course of
therapy and the adverse drug reactions and interactions that may follow
(WALLACE and WATANABE 1977; OUSLANDER 1981; BRAVERMAN 1982;
GREENBLATT et al. 1982; SHAW 1982; D'ARCY and McELNAY 1983; ROYAL
COLLEGE of PHYSICIANS 1984; NOLAN and O'MALLEY 1988a,b; DENHAM 1990;
WILLIAMS and LOWENTHAL 1992). Of these factors, age and genetic influences
have been pinpointed as significant contributors to problems with drug
therapy.
This chapter deals with these two influences in turn: Sect. B, "Age", and
Sect. C, "Genetic Factors", and it will highlight some of their more important
influences on drug action. Where possible, it will indicate the mechanisms that
underline these influences.
B. Age
It has long been established that elderly patients use more medicaments than
younger age groups (LANDAHL 1987; WILLIAMS and LOWENTHAL 1992; SLOAN
1992; STEWART and COOPER 1994) and thus have a greater risk of drug-drug
interactions occurring (KELLAWAY and MCCRAE 1973; LAWSON and JICK 1976;
LEVY et al. 1980; WILLIAMSON and CHOPIN 1980; SIMONS et al. 1992; SCHENKER
and BAY 1994; STANTON et al. 1994; STEWART and COOPER 1994) (Fig. 1). It must
be understood, however, that there is virtually no direct evidence in the
literature that age per se can cause drug-drug interactions. Its role is more in
enhancing the effects of drug-drug interactions when they occur. Age thus
tends to exert a quantitative influence on the interaction but does not alter its
qualitative spectrum.
Senescence is frequently evoked to explain the unwanted sequelae to
therapy, undoubtedly rightly - provided senescence is regarded as being ac-
companied, for example, by physiological changes due to age (LAMY 1991), by
small body mass, poor renal function, and impaired function of other organs,
notably the liver. Table 1 shows some of the key changes that occur as a result
of physiological ageing. In elderly patients the reserve capacity of many organs
may be considerably reduced, and because of this erosion there is a narrowing
280 J.e. McELNAY and P.F. D'ARCY
VI 100
U
.!!!
Q;
., 80
.,l!.'
>
"0
co
£: 60
'ji;
VI
C 40
.!!!
iii
.,
0.
Ol
co 20
.,u
C
Q;
[l..
2 3 4 5 6 7 8 9 10
Number of drugs
Fig. 1. Relationship between percentage of patients with ADR and the number of
drugs prescribed. (DENHAM 1990)
of the safety margin between the therapeutic and toxic dose of many drugs. As
a result of this the elderly, as a group, get rather more than their fair share of
drug-induced disease and the complications of drug-drug interactions. This
especially applies to psychogeriatric patients in whom the nature of drugs, the
number used concomitantly, and the long-term use presents peculiar hazards.
Table 2 shows, as an example, the drug-drug interactions identified by
GOSNEY and TALLIS (1984) in their survey of interacting drugs in elderly
patients admitted to hospital. Nine of these combinations were potentially life-
threatening; 51 combinations were likely to lead to potentially serious side
effects, and 27 were significant in as much as they could lead to suboptimal
treatment. Other accounts of common drug-drug interactions are given
throughout this volume and will not be repeated here. It is sufficient to
emphasise that qualitatively the elderly appear to suffer the same adverse
effects as younger age groups but they suffer them at an enhanced level.
Many adverse effects of drugs or drug combinations that may simply be a
nuisance to younger patients are much intensified in the elderly due, for
example, to their decreased resistance to the adverse consequences. Indeed,
the adverse effects of drug treatments may convert the elderly patient from a
functional sentinent human being into a chair-fast incontinent wreck.
In their simplest form, drug-drug interactions cause either an enhanced
pharmacological or toxicological effect of one or more of the combination
drugs, or conversely a reduced therapeutic effect of one or more of the com-
ponents. A number of investigations have shown that age is not an indepen-
dent risk factor (NOLAN and O'MALLEY 1989; CARBONIN et al. 1991; GURWITZ
and AVORN 1991; CHRISCHILLES et al. 1992). However, regardless of whether
the elderly are more sensitive to the adverse effects of medication, they have
more disease, consume more medications and are overrepresented in nearly
every study of adverse drug reactions (SCHNEIDER et al. 1992).
Age and Genetic Factors in Drug Interactions 281
Table 1. Some key changes as a result of physiological aging. (COLLIER et al. 1993)
tv
00
(;>
284 J.e. McELNAY and P.F. D' ARCY
1. Absorption
The extent of drug absorption is generally little affected by age. It is true that
physiological changes occur with ageing that can influence drug absorption, for
example, decreased gastric acid secretion, decreased pancreatic lipase activity,
decreased gastric emptying and gastrointestinal motility, and decreased gastric
blood flow. However, although delays in absorption have been described for
some substances, they are generally of little clinical importance since the
extent of absorption remains largely unchanged. The absorption of levodopa
is an exception. It has been demonstrated, for example, that there can be a
threefold increase in its absorption in elderly patients probably due to an age-
related reduction in gastric dopamine decarboxylase and consequently greater
availability of levodopa for absorption into the systemic circulation (EVANS et
al. 1980). This increase in absorption is probably one of the main reasons for
the substantially lower dose of levodopa required for elderly patients (BROE
and CAIRD 1973) and for levodopa being one of the commoner causes of
adverse drug reactions (ADRs) in elderly patients.
2. Distribntion
Several blood flow changes occur with advancing years. Firstly there is a
decrease in cardiac output of 1 %/year after the age of 25 years. Secondly, in
older adults there is a decreased perfusion of limbs, liver and mesentery of up
to 45%. Thirdly, in elderly people there is a decreased blood flow to the heart
Age and Genetic Factors in Drug Interactions 285
(30%) and brain (15 %). Although little data are available, these changes in
blood flow could obviously influence the rate of drug distribution to various
tissues. In the elderly, however, it is the changes in body composition, includ-
ing the concentration of plasma albumin, which have the greatest impact on
drug distribution.
In the elderly, lean body mass is reduced, fat mass is increased and intra-
cellular fluid volume is decreased. The implications of these changes for drug
kinetics will depend largely on the degree of lipid and aqueous solubility of the
drug in question and its degree of binding to plasma albumin. The volume of
distribution of lipid-soluble drugs, e.g., diazepam and lignocaine, will obvi-
ously be increased while the opposite effect will be seen with water-soluble
drugs, e.g., gentamicin and nadolol. These effects will tend towards lower
plasma concentrations of lipid-soluble drugs and higher concentrations of
water-soluble drugs. Lipid-soluble drugs, as well as distributing into body fat,
can concentrate in the brain, which is high in lipoid tissue. There is also some
evidence to suggest that the blood-brain barrier is less intact in the elderly,
thus allowing drugs to distribute into the brain in increased concentrations.
Changes in drug distribution are of particular significance when the therapeu-
tic ratio of the drug is low as in the case of digoxin, where the 30% reduction
in its volume of distribution in the elderly is probably related to reduction in
lean body mass. This necessitates a reduction in the loading dose of digoxin in
elderly patients.
For drugs which are highly bound to plasma proteins, the decreased
concentrations of albumin that occur, particularly in sick or malnourished
elderly patients, can also have marked effects on drug distribution and there-
fore drug plasma levels. As a general rule, drugs which are highly bound to
plasma proteins distribute very little into organs since bound drug cannot cross
the capillary membranes, due to the large size of the protein molecules.
Warfarin is a good example of this effect - it is highly bound to plasma albumin
(>99%) and is largely retained in the plasma compartment. In the elderly,
however, since albumin concentrations are decreased, less drug is bound and
more drug is free to distribute throughout the body and to be eliminated by the
liver and kidney. This redistribution leads to decreased total concentrations
(free + bound) of drug in the plasma, but, in the absence of effects on drug
elimination, one would expect the free, pharmacologically active drug concen-
trations to remain relatively similar. In the elderly, however, drug elimination
is often decreased and therefore the free drug concentrations in plasma may
become elevated with consequent increases in pharmacological effects and
toxicity. When drugs are bound to albumin there is also a greater risk of
displacement due to interaction between drugs competing for the same bond-
ing sites if the serum albumin levels are reduced (see Chap. 4). In contrast to
albumin, plasma globulin concentrations tend to increase with increasing age.
This effect is relatively unimportant since few drugs are extensively bound to
these proteins.
286 J.e. McELNAY and P.F. D'ARCY
Some drugs are taken up actively into various tissues, particularly if they
resemble a naturally occurring nutrient. This method of tissue uptake is
utilised by some of the antitumour drugs, for example, the uptake of
melphalan, an amino acid derivative, into tumour cells has been shown to
involve active transport. One might expect the active uptake of drugs into
tissues to be decreased in the elderly due to declining metabolic function;
however, good data are not available to support this hypothesis.
In summary, the main effects of ageing on drug distribution include a
more widespread distribution of lipid-soluble drugs, more limited distribution
of water-soluble drugs and a more extensive distribution of drugs bound to
plasma albumin. A summary of the effects of ageing on drug distribution is
presented in Table 3 (RITSCHEL 1980).
Table 3. Continued
Methotrexate t J- J-
Metoprolol t NI NI
Morphine t NI NI
Netilmicin t U J-
Nitrazepam t U
Nortriptyline t U J-
Oxazepam t t NI
Penicillin G t NI NI
Phenobarbital t NI NI
Phenylbutazone t U J-
Phenytoin NI NI t
Procaine penicillin t NI NI
Practolol t J- NI
Propicillin t J- NI
Propranolol t J- J-
Protriptyline t J- J-
Quinidine t J- J-
Spironolactone t NI NI
Sulbenicillin t t NI
Sulfamethizole (sulphasomidine) t U or J- J-
Tetracylline t NI NI
Theophylline t U or J- J-
Thioridazine t NI NI
Tobramycin t J- J-
Tolbutamide t J- J-
Warfarin t U J-
3. Metabolism
Since the early animal experiments of CONNEY et al. (1956, 1957) and CONNEY
(1967) it has often been observed that the rate of oxidative metabolism of
various substrates can differ markedly depending on a number of factors of
which age is important. However, direct measurement of enzyme levels in liver
biopsy samples has shown few age-related alterations in the concentration of
several drug-metabolising enzymes in man (WOODHOUSE et al. 1984). Human
studies have suggested that the changes observed in clearance of oxidatively
metabolised drugs are more likely due to physiological changes in elderly
patients than to changes in their cytochrome P450 systems. There are few, if
any, detectable changes related to ageing in cytochrome P450 protein or
mRNA (SCHMUCKER and KONTAK 1990). Age-related reduction in overall liver
mass and absolute liver blood flow are most likely the important factors in
diminishing drug metabolism (CALLOWAY et al. 1965; SKAUNIC et al.1978). Age
may affect hepatic oxidative processes to a greater extent than conjugative
metabolic processes (see Table 4). Although an individual's liver function is
difficult to assess, problems are more likely in patients with ascites, jaundice
and encephalopathy.
288 J.e. McELNAY and P.F. D'ARCY
Table 4. Studies on the relation of age to the clearance of drugs by hepatic bio-
transformation. (GREENBLATI et al. 1982)
4. Excretion
The two main routes by which drugs are eliminated from the body are metabo-
lism by liver enzymes, and excretion by the kidneys. The chemical changes
that result from liver metabolism generally (but not always) result in mol-
ecules that are less active pharmacologically and are less lipid soluble and/or
more easily ionised. This causes them to be more readily excreted by the
kidneys.
Age and Genetic Factors in Drug Interactions 289
drugs were identified in 200 (3.2%) of 6160 prescriptions. One hundred and
thirty-six (23.7%) patients were affected. The most common interactions (po-
tential or actual) were frusemide with aminoglycosides (increased ototoxicity);
cimetidine with antacids (interference with action/absorption); frusemide with
prednisolone (potassium loss); loop diuretics with NSAIDs (antagonism
of diuretic effect), and frusemide with cephalosporins (increased
nephrotoxicity).
NOLAN and O'MALLEY (1989) evaluated the risk of potential drug inter-
actions in 11 private nursing homes in Dublin. The percentage of patients
prescribed potentially interacting combinations increased greatly with the
number of medications taken.
Potential drug-drug interactions were also studied in an ambulatory popu-
lation in Florida (HALE et al. 1989). Ten major interaction classifications were
studied. It was found that 40% of patients taking quinidine were also taking a
digitalis glycoside, and nearly one-third of patients taking warfarin were also
taking a drug with an interacting potential.
SCHNEIDER et al. (1992) investigated adverse drug reactions in an elderly
outpatient population attending an interdisciplinary geriatric clinic and a
medical clinic in Cleveland, Ohio, United States. The sample size of the study
was 463 patients, of whom 332 attended the medical clinic and 131 attended
the geriatric clinic. Potential drug interactions were identified in the records of
143 (31 %) sUbjects.
DOUCET et al. (1993), from Rouen and Bois Guillaume, studied drug
interactions in French patients over 65 years old. Of 513 elderly patients
admitted to hospital, the principal drug interactions occurred with diuretics
and benzodiazepines. Of this population 63 % had one or more drug interac-
tions leading to an adverse effect (124 patients) and to hospitalisation in half
these cases.
STANTON et al. (1994) studied drug-related admissions to an Australian
hospital. The median age of the patients was 67 years; 4.4 % of admissions were
due to drug interactions. SCHENKER and BAY (1994) working in a Veterans
Medical Administration Center in Texas, United States, investigated drug
disposition and hepatotoxicity in the elderly and concluded that drug-drug
interactions and concurrent derangements accompanying advanced age made
a significant contribution to adverse drug effects.
Apart from these group studies, there have been a large number of single
patient reports in which elderly patients suffered drug interactions. For ex-
ample, KERR (1993) in the United States described an elderly woman who
developed gastrointestinal bleeding probably potentiated by the interaction of
fluconazole with warfarin. SCARFE and ISRAEL (1994) also in the United States,
reported the case of an elderly woman who experienced a significant increase
in INR after levamisole and fluorouracil were added to an established regimen
of warfarin.
These studies confirm the obvious, that as the number of medications in a
patient's regimen increases, the potential for interacting combinations also
Age and Genetic Factors in Drug Interactions 293
V. Long-Term Treatments
Most of the drugs involved in clinically relevant interactions are those on
which patients are carefully stabilised for relatively long periods (e.g., oral
294 J.e. McELNAY and P.F. D'ARCY
c. Genetic Factors
The term "pharmacogenetic disorder" was originally coined by VOGEL (1959)
and was originally limited to hereditary disorders revealed solely by the use of
drugs. Its meaning has now been enlarged to embrace all genetic contributions
to the considerable variation that exists in the interaction between man and
the pharmacological agents that he uses. The term can therefore be taken to
cover the adverse drug reactions and interactions that occur when particular
drugs are given to a patient with a genetically determined dysfunction of an
organ or body system, for example, renal polycystic disease.
As with the effect of age on the incidence or severity of drug-drug interac-
tions, there is no reason to suggest that genetic influences cause drug interac-
tions per se. What is clear, however, is that patients who suffer the effects of
pharmacogenetic disorders may well show a predisposition to specific drug-
drug interactions, or may show enhanced effects when such interactions occur.
The risk of drug toxicity usually arises from enzyme deficiency states.
Examples of these have been well described by BENNETI (1993). For example:
Hepatic porphyrias, where deficient conversion of porphyrins to haem
exposes affected individuals to risk from drugs which have the common prop-
erty of increasing the activity of delta-aminolaevulinic acid synthetase, the
rate-limiting enzyme of porphyrin synthesis, in their livers. Disorders of por-
phyrin metabolism have been comprehensively reviewed by FLETCHER and
GRIFFIN (1986) and they have presented a figure of the overall scheme of
porphyrin synthesis and a list of those drugs which have been recognised as
being associated as precipitating agents of porphyria. The figure (Fig. 2) and
list of drugs (Table 5) are reprinted here.
Malignant hyperpyrexia is a rare pharmacogenetic disease (rigid and non-
rigid types) occurring both in man and in the Landrace strain of pigs. In
susceptible individuals any potent inhalation anaesthetic or any skeletal
muscle relaxant can cause fever, rigidity, hyperventilation, cyanosis, hypoxia,
'INHERITED DISORDER
Biochemical detect in porphyria believed
Acetylating mechanisms to be an excessive activity of ALA synthetase,
(Acetylchotine) the rate controlling enzyme of the synthesis.
Various forms of disease probably due to
~
(l)
disturbances lower down chain resulting
ACEm.COA< from overactivity of ALA synthelase. §
0..
Other melabolic pathways /
C'l
(l)
presence of ~
(l)
1
SUCCINYL co A. + GLYCINE -----.. a AMINO-f}OXO ADIPIC ACID O.
(')
pyridoxal
phosphate loss of C02 under influence of ALA synthetase
1 "TI
~
(')
1 condensation 8'
PORPHOBILINOGEN (PBG) I>-AMINO LAEVULINIC ACID (ALA) ....
of two molecules '"5'
under influence
of aminolaevutate o
dehydratase
Four molecules of 1 &
PBG under influence INTERMEDIATE PRODUCTS ......
~
of porphobilinogenase Uroporphyrinogen I Uroporphyrin I /urine 100)1g porphyrin/day rl
• (l)
combine 0.7mg ....
formed (TYPE 1) TYPE 1 ~
~ perday ~
Coproporphyrinogen I Coproporphyrin I o·
• \ I '\. ,,,'~ "'"'" ".",rio"" ~
1 '"
dehydrogenation EXCRETED
UROPORPHYRINOGEN III Uroporphyrin III Urine 50)1g porphyrin/day
PROTOPORPHYRIN r Haemoproteins t
Haemogiobin ~ Cytochromes
Myoglobin
t5
VI
Table 5. Drugs classified as to their association with porphyria. (FLETCHER and GRIFFIN
1986)
Table 5. Continued
Potentially Drugs believed not Potentially Drugs believed not
porphyrogenic drugs to precipitate porphyrogenic drugs to precipitate
porphyria porphyria
Pethidine Primaquine Spironolactone Tubocurarine
Phenazone Propoxyphene Steroids Sodium valproate
Phenelzine Propranolol Streptomycin
Phenoxybenzamine Prostigmine Succinimides Vitamin C
Phensuximide Pyrimethamine (ethosuximide,
Phenylbutazone methsuximide,
Phenylhydrazine Quinine phensuximide)
Primidone Sulphonal
Probenecid Reserpine Sulphonamides (A)
Progestogens (A, C) Resorcinol Sulphonylureas
Propanidid Rifampicin Sulthiame
Pyrazinamide
Pyrazolones Streptomycin Tetracyclines
(antipyrine, Succinylcholine Theophylline
isopropylantipyrine, Tolazamide
dipyrone, sodium Tetracyclines Tolbutamide (A, C)
phenyl dimethyl Tetraethylammonium Tranylcypromine
pyrazolone) bromide Trional
Pyridoxine Thiouracils Troxidone
Pyrimethamine Tricyclic
antidepressants Valproic acid
Ranitidine (amitriptyline) Viloxazine
Rifampicin Trifluoperazine
Thiazides Xylocaine
Sodium valproate Tripelennamine
Drug Haemolysis
Acetanilide +++++
Dapsone ++ +++
Furazolidone ++
Nitrofural ++++
Nitrofurantoin ++ ++
Sulphanilamide +++
Sulphapyridine +++ +++
Sulphacetamide ++
Salazosulphapyridine +++
Sulphamethoxypyridazine ++
Thiazosulphone ++
Quinidine ++
Primaquine +++ +++
Pamaquine ++++
Pentaquine +++
Quinocide +++ ++
Naphthalene +++ +++
Neoarsphenamine ++
Phenylhydrazine +++
Toluidine blue ++++
Trinitrotoluene +++
300 J.e. McELNAY and P.F. D' ARCY
D. Comment
This chapter is not able to show that age or genetic make-up is directly
responsible for drug-drug interactions. The examples of therapeutic hazard
that have been given in this text are largely those due to adverse drug reac-
tions. It has been shown, however, that both age and genetic factors can
seriously affect drug therapy either by reducing its effect or alternatively
enhancing its effect, including increased toxicity.
It has been shown in many studies that the elderly as a group have to take
more medicines than younger age groups because of the disease conditions
Age and Genetic Factors in Drug Interactions 301
that occur as age progresses. It is also a well-established fact that the more
medicines that are taken together the greater the possibility of drug-drug
interactions occurring. It therefore follows that the elderly are likely to suffer
the effects of more drug-drug interactions than younger age groups and that,
due to decreased resilience in the elderly, such interactions will probably have
greater effect on their continued health than similar interactions would have
on younger patients.
Genetic abnormalities can increase the toxicity of drug therapy at normal
therapeutic dosage levels. This may therefore increase the possibility of hazard
from specific drug-drug interactions. Medical opinion does not generally
favour multiple-drug therapy, but on occasion it is necessary. The price that
may be required for using multiple drugs in elderly patients and in those
persons with an abnormal genetic make-up may be the occurrence of relatively
more and more serious drug-drug interactions.
References
Adams KRH, Al-Hamouz S, Edmund E, Tallis RC, Vellodi C, Lye M (1987) Inappro-
priate prescribing in the elderly. J R Coll Physicians Lond 21:39-41
Agate J (1963) The practice of geriatrics. Heinemann, London, pp 217-222
Armaly MF (1968) Genetic factors related to glaucoma. Ann NY Acad Sci 151:861-
875
Baker SP, Harvey AH (1985) Fall injuries in the elderly. Clin Geriatr Med 1:501-512
Bennett PN (1993) Evaluation of toxicity in human subjects. In: Ballantyne B, Marrs T,
Turner P (eds) General and applied toxicology, voll. Macmillan, Basingstoke, pp
367-384
Bourne JG, Collier HOJ, Somers GF (1952) Succinylcholine (succinoylcholine)
muscle-relaxant of short action. Lancet 1:1225-1229
Bovet D, Depierre F, Courvoisier S, de Lestrange Y (1949) Synthetic curarizing agents.
II. Phenolic ethers with quaternary ammonium groups. The action of tris
(diethylamino-ethoxy) benzene triiodoethylate (2559F). Arch Int Pharmacodyn
80:172-188
Braverman AM (1982) Therapeutic considerations in prescribing for elderly patients.
Eur J Rheumatol Infiamm 5:298-293
Britt BA, Kalow W (1968) Hyperrigidity and hyperthermia associated with
anaesthesia. Ann NY Acad Sci 151:947-958
Britt BA, Locher WG, Kalow W (1969) Hereditary aspects of malignant hyperthermia.
Can Anaesth Soc J 16:89-98
Broe GA, Caird PI (1973) Levodopa for parkinsonism in elderly and demented pa-
tients. Med J Aust 1:630--635
Caird FI (1985) Towards rational therapy in old age. The F.E. Williams Lecture 1985.
J Roy Coll Physicians Lond 19:235-239
Calloway NO, Foley CF, Lagerbloom P (1965) Uncertainties in geriatric data. II.
Organ size. J Am Geriatr Soc 13:20-28
Carbonin P, Pahor M, Bernabei R et al. (1991) Is age an independent risk factor of
adverse drug reactions in hospitalized medical patients? J Am Geriatr Soc
39:1093-1099
Carpopresso PR, Gittleman MA, Reilly DJ, Paterson LJ (1975) Malignant
hyperthermia associated with enfiurane anaesthesia. Arch Surg 110:1491-1493
Chrischilles EA, Segar ET, Wallace RB (1992) Self-reported adverse drug reactions
and related resource use. A study of community-dwelling persons of 65 years of
age and older. Ann Intern Med 117:634-640
302 J.e. McELNAY and P.F. D' ARCY
Schneider JK, Mion LC, Frengley JD (1992) Adverse drug reactions in an elderly
outpatient population. Am J Hosp Pharm 49:90-96
Schwartz JT, Reuling FH, Feinlieb M, Garrison RJ, Collie DJ (1972) Twin heritability
study of the effect of corticosteroids on intraocular pressure. J Med Genet 9:137-
143
Scott PJ, Reid JL (1982) The effect of age on the response of human isolated arteries
to noradrenaline. Br J Clin Pharmacol 13:237-256
Shaw PG (1982) Common pitfalls in geriatric drug prescribing. Drugs 23:324-328
Shephard AM, Hewick DS, Moreland TA, Stevenson IH (1977) Age as a determinant
of sensitivity to warfarin. Br J Clin Pharmacol 4:315-320
Simons LA, Tett S, Simons J, Lauchlan R, McCallum J, Friedlander Y, Powell 1(1992)
Multiple medication use in the elderly - use of prescription and nonprescription
drugs in an Australian community setting. Med J Aust 157:242
Skaunic V, Hulek P, Martinkova K (1978) In: Kitani K (ed) Liver and ageing. Elsevier/
North Holland, Amsterdam, pp 115-130
Sloan RW (1992) Principles of drug therapy in geriatric patients. Am Fam Physician
45:2709-2718
Smith RL, Idle JR (1981) Genetic polymorphism in drug oxidation. In: Davis M,
Tredger JM, Williams E (eds) Drug reactions and the liver. Pitman, Bath, pp 95-
104
Stanton LA, Peterson GM, Rumble RH, Cooper GM, Polack AE (1994) Drug-related
admissions to an Australian hospital. J Clin Pharmacol Ther 19:341-347
Stewart RB, Cooper JW (1994) Polypharmacy in the aged. Practical solutions. Drugs
Aging 4:449-461
Sutherland FS, Carter JR (1975) Malignant hyperpyrexia during enflurane anaesthesia.
J Tenn Med Assoc 68:785-786
Telfer ABM, Macdonald DJZ, Dinwoodie AJ (1964) Familial sensitivity to
suxamethonium due to atypical pseudocholinesterase. Br Med J 1:153-156
Tinetti ME, Speechley M, Ginter SF (1988) Risk factors for falls among elderly persons
living in the community. N Engl J Med 319:1701-1707
Vesta RE, Wood AJ, Shand DG (1979) Reduced beta-adrenoreceptor sensitivity in the
elderly. Clin Pharmacol Ther 26:181-186
Vogel F (1959) Modem problems of human genetics. Ergeb Inn Med Kinderheilkd
12:52-125
Wallace DE, Watanabe AS (1977) Drug effects in geriatric patients. Drug Intell Clin
Pharm 11:597-603
Williams L, Lowenthal DT (1992) Drug therapy in the elderly. South Med J 85:127-131
Williamson J, Chopin JM (1980) Adverse reactions to prescribed drugs in the elderly:
a multicentre investigation. Age Aging 9:73-80
Woodhouse KW, Mutch E, Williams FM, Rawlins MD, James OF (1984) The effect of
age on pathways in drug metabolism in the human liver. Age Ageing 13:328-334
World Health Organization (1981) Health care in the elderly: report of the technical
group on the use of medicaments by the elderly. Drugs 22:279
CHAPTER 11
Drugs Causing Interference
with Laboratory Tests
S. YOSSELSON-SUPERSTINE
A. Introduction
Laboratory tests, along with the patient's history and the physical examina-
tion, often provide the main key to accurate diagnosis. In some cases their
abnormality is the only clue to diagnosis. Results of laboratory tests are also a
guide to rational therapy. They can reflect the effectiveness of the therapeutic
agent employed as well as indicate the appearance of adverse reactions. No
proper diagnosis and therapy can be provided without accurate and reliable
laboratory tests. Thus the sensitivity and the specificity of the test method
should be among the most important factors, more than cost and ease of
performance, in choosing and adopting a method or equipment. Specificity is
defined as the proportion of the true negatives that are correctly identified by
the test (ALTMAN and BLAND 1994) and is affected inversely by the number of
false-positive measurements as shown in the following formula (GADDIS and
GADDIS 1990):
CIty (0//0 ) =
SpecI·fi· True negatives x 100
True negatives + false positives
Many factors can cause a laboratory error or false-positive results. They
include human error, equipment or environmental changes, chemicals added
to specimens and the presence of endogenous substances. Another important
factor which has an effect is the presence of medications in the fluid tested
or influencing the functions in the body; this is then reflected in the test result.
This last factor - medications - is often overlooked. The mechanism of
their interference is the subject of this review. It is not the purpose of
this chapter to survey the literature and summarize the drug interferences
which have been documented with the various laboratory tests, nor is it
the aim to establish the clinical significance of drug-test interference. Only
examples of important interferences representing different mechanisms
will be discussed in more detail. The design of a study for the determination of
a clinically significant drug-test interaction resulting from methodological
interference will also be described. The reader is referred to excellent
compilations of published studies on this topic (SALWAY 1990; YOUNG 1990)
and to critical evaluations of some of the relevant literature (YOSSELSON-
SUPERSTINE 1986, 1989).
306 S. YOSSELSON-SUPERSTINE
I. Pharmacological Interferences
Pharmacological interferences are by far the most frequent type of interfer-
ence. They affect the result of the test by virtue of the activity of the drug or
its metabolites in the human body, regardless of the method employed in the
test. A pharmacological effect of a drug on a laboratory test is easy to detect
when the change in the test value is expected and wanted, but much more
difficult to interpret when it is an unexpected, toxicological affect of the drug.
The following examples demonstrate these effects:
effect in as many as half the patients receiving them (ANON 1980). The preva-
lence of asymptomatic hyperuricemia could even rise to 65%-75% in elderly
patients being treated with diuretic, while the development of symptomatic
gout is commonly seen in only 1 %-2% of these individuals. Uric acid retention
begins soon after diuretic therapy is started, is dose dependent and produces
an average increase in serum uric acid concentration of 1.2-1.5 mgldl (WADE et
al. 1989). The precise mechanism is unclear. Several mechanisms have been
suggested, among them increased proximal tubular renal reabsorption, de-
creased tubular secretion and increased post-secretory reabsorption of uric
acid (MAY et al. 1992). The increase in uric acid will be demonstrated by all
laboratory methods for uric acid measurement, regardless of the technique
employed, further proof that the interference is pharmacological or toxicologi-
cal in its nature.
first described was even thought to be an interference with the digoxin assay
methodology, which, if true, would not have necessitated an adjustment in the
digoxin dosage. However, this possibility was excluded by measuring digoxin
in plasma of patients on quinidine alone and finding no measurable concentra-
tions and by looking for an effect of quinidine on digoxin levels while adding
both drugs to plasma in vitro (EJVINSSON 1978).
1. Colorimetric Interferences
Most of the laboratory tests in chemical pathology still have a colorimetric
component, which could be subject to the effect of foreign chromagens. The
following are examples of such interferences:
tion was noticed when the method of HENRY et al. (1957) was employed. This
method used carbonate-phosphotungstate and eliminated the use of cyanide
in the reaction used previously in order to reduce the turbidity of the final
colored solution, which made colorimetric measurement difficult. In addition
to modifying the test to eliminate acetaminophen interference, SINGH et al.
were also able to demonstrate a log-linear relationship between the concentra-
tion of the drug in aqueous solution and the concentration of the apparent uric
acid, further suggesting that the interference was with the method of uric acid
determination and not merely a pharmacological effect of the drug on uric
acid. CARAWAY (1969) demonstrated that ascorbic acid in a concentration of
IO.ug/ml, which could be found in the blood after the consumption of 2g of
the vitamin, increased serum uric acid concentration by 1.34mg/dl in
pooled sera and in sera of 21 patients, when determined by a carbonate-
phosphotungstate method. This interference is not significant in the more
sensitive cyanide methods but is of importance in methods where cyanide
is replaced by sodium silicate or carbonate (ALPER and SEITCHIK 1957).
Ascorbic acid was eliminated by mild alkaline treatment prior to adding
phosphotungstic acid (CARAWAY 1963). Incubation for 10min after adding
sodium carbonate effectively destroys the ascorbic acid present in serum and
urine in physiological concentrations.
High concentrations of uricase-resistant chromagens have been reported
in the serum of gouty patients receiving high maintenance doses of salicylate
(GRAYZEL et al. 1961). These metabolites have not been identified but were
not found to be gentisic acid - a salicylate metabolite interfering with uric acid
colorimetric assay in urine but not in plasma (CARAWAY 1969). The plasma
levels of salicylates in GRAYZEL'S study were above 13mg/lOOml. It should be
noted that at these levels there is also a pharmacological interference, a
uricosuric effect of salicylates, resulting in a lowering of uric acid levels. This
can affect the 7%-119% rise in uric acid as measured by a colorimetric method
(YO and GUTMAN 1959; GRAYZEL et al. 1961). On the other hand, the rise in
uric acid levels at salicylate doses of less than 2g/day is again caused by a
pharmacological effect of the drug leading to urate retention. Unconjugated
salicylate in the renal tubule may act by blocking a postulated tubular
secretion (YO and GUTMAN 1959). The distinction between the two types of
interferences - pharmacological and methodological - was possible by
measuring uric acid twice, once by the colorimetric assay and once by an
enzymatic spectrophotometric assay (GRAYZEL et al. 1961). These interfer-
ences are of great clinical significance since they can lead to an erroneous
diagnosis of gout in rheumatoid arthritic patients receiving various doses of
aspirin containing medications.
CAWEIN and HEWINS (1969) have demonstrated a 20% increase in serum
uric acid in 18 patients who received 3-7 g levodopa daily. This elevation was
absent when the uricase method was used. However, a pharmacological effect
cannot be ruled out since an elevation in serum uric acid, measured by the
uricase method, in two patients (AL-HuJAJ and SCHONTHAL 1971, 1972) and
310 S. YOSSELSON-SUPERSTINE
b) Creatinine Analysis
Creatinine measurement is one of the most valuable laboratory tools in clinical
practice. Not only does it provide an excellent estimation of the patient's
kidney function, but it also serves as a guide to dosage adjustments of drugs
whose elimination is mainly by the renal route. Two major techniques are used
today for the analysis of creatnine: colorimetric and enzymatic. The colorimet-
ric method is based on Jaffe's reaction, which was developed more than a
century ago and is still the most common assay for the determination of
creatinine either in serum or in urine (JAFFE 1886). It is employed in many
reagent kits and instruments, such as Beakman ASTRA-8, Technicon SMAC
and Du Pont ACA. In this reaction there is development of a red color
when creatinine reacts with picric acid in an alkali environment. The colored
end product absorbs light between 490 and 520nm and is measured by
spectrophotometer. The Jaffe reaction is efficient and inexpensive, but is
influenced not only by environmental conditions, and the amount of the
chemicals used, but also by noncreatinine chromagens, which could account
for 20% of the total measured creatinine in serum (NARAYANAN and ApPLETON
1980). These chromagens could be physiologic substances such as
glucose or protein (DATTA et al. 1986), as well as medications such as some of
the cephalosporin (GROTSCH and HAJDU 1987) and penicillin antibiotics
(KROLL et al. 1984a), lactulose (BRUNS 1988), high-dose furosemide infusion
(MURPHY et al. 1989), the today rarely used methyldopa (MADDOCKS et al.
1973; NANJI and WHITLOW 1984), acetohexamide (ROACH et al. 1985) and
phenacemide (RICHARDS 1980). Of the many cephalosporins studied, only
cefoxitin and cephalothin produced a significant interference at therapeutic
concentrations of 100mg/l (GREEN et al. 1990), as well as cafazolin, which
caused a false increase of 10-20,umol/l (0.1--D.2mg%) of creatinine for every
20mg/l cefazolin (NANJI et al. 1987).
The many publications on the interferences of these medications, and
especially cephalosporins, with the Jaffe analysis were evaluated very exten-
sively in a recent review (DUCHARME et al. 1993). The mechanism of interfer-
Drugs Causing Interference with Laboratory Tests 311
or brown urine suggests hematuria, and orange or dark yellow urine suggests
the presence of urobilin. Bleeding from the upper gastrointestinal tract may
cause the stool to be black.
Drugs excreted in urine or feces can change the normal color and appear-
ance of these body wastes and obscure the elementary gross examination of
these specimens. Some also have the potential to affect tests performed on
urine or feces, especially when they are based on colorimetric methods.
Therefore, before proceeding with other diagnostic measures, a detailed
history of dietary intake of food and drugs should be obtained from the
patient. For instance, orally administered iron can cause false-positive reac-
tions for fecal occult blood when tested by the Hemoccult and Hematest
methods (LIFTON and KREISER 1982). Hemoccult uses guaic as a reagent and
Table 1. Continued
fiector photometers that could read the reagent strips, allowing for elimination
of the variable that has commonly caused erroneous and confused results
when color has been interpreted by the human eye (ANON 1982).
Highly buffered, alkaline urines may give false-positive results when the
buffer systems in the reagent area are overcome and an actual shift in pH of
the buffers occurs. Addition of sodium carbonate (43 gil urine standard)
caused a +4 reading by the Albustix method (GYURE 1977).
Therapeutic doses of the commonly used antacids in over the counter
medications do not elevate urine pH by more than one unit (GIBALDI et al.
1974). Since normal pH varies from 4.6 to 8, it is unlikely that they affect urine
protein determination by the reagent strip method unless, at least theoreti-
cally, there is an abuse of antacid products.
Therapeutic doses of acetazolamide (37S-1000mg/day) have also not been
found to elevate urine pH to 10 or above and did not falsely elevate urine
protein levels (YOSSELSON-SUPERSTINE and SINAI 1986).
a) Creatinine Analysis
The enzymatic method for creatinine determination, which is used in the
automated drug-slide system, is based on the enzymatic hydrolysis of creati-
nine by creatinine iminohydrolase and the production of ammonia and N-
methylhydantoin. The ammonia reacts with bromophenol blue to form a blue
chromagen, the amount of which is measured spectrophotometrically. Its
concentration thus reflects the concentration of creatinine (TOFFALETII et al.
1983).
The 4-amino group of flucytosine, a systemic antifungal drug, can be
converted to ammonia by the same enzymatic system and cause false elevation
318 S. YOSSELSON-SUPERSTINE
a) Digoxin Assay
Digoxin is one of the most important drugs where the therapy is guided by its
concentrations in the patient's blood. All the immunoassays, radioimmunoas-
says, enzyme immunoassays and fluoroimmunoassays used for measurement
of the drug suffer from the cross-reactivity of digoxin metabolites and from
endogenous substances known as digoxin-like immunoreactive substances
(DLIs). These substances accumulate in the blood, especially in neonates,
during the third trimester of pregnancy, renal failure and liver disease (MORRIS
et al. 1990). The most important exogenous substance to interfere with digoxin
assays is the diuretic spironolactone - a synthetic steroid. Canrenone and its
20-hydroxy derivative, both metabolites of spironolactone, have apparently
more cross-reactivity with digoxin than the parent drug (HUFFMAN 1974;
SILBER et al. 1979; MORRIS et al. 1987). The wide range of falsely increased
levels of digoxin ranging from no effect to an increase of 4.0nglml after the
administration of therapeutic doses of spironolactone (25-200mg/day) can be
attributed to the difference in the specificity of the digoxin antiserum used in
the different RIA kits (MORRIS et al. 1987).
Endogenous steroids are known to cross-react with digoxin. An apparent
digoxin concentration of 0.5 mg/l has also been reported in the serum of a
patient receiving no digoxin, 30min after the administration of 6-methylpred-
nisolone (GAULT et al. 1985). The interference with steroids was noted with
both radioimmunoassay and with fluorescence polarization immunoassays
(SOLDIN et al. 1984). Fab fragments infused into patients with digoxin toxicity
pose another problem with digoxin radioimmunoassays, as well as with
fluorescence excitation transfer immunoassays (NATOWICZ and SHAW 1991).
Digoxin serum levels after Fab treatment could be as much as tenfold greater
than pretreatment values, when the solid-phase methods are used. The inter-
ference could be eliminated when polyethylene glycol and charcoal methods
320 S. YOSSELSON-SUPERSTINE
are used (GIBB et al. 1983). It is best not to use digoxin blood levels as a
guideline for continuation of the antidote therapy and to base the decision on
the clinical presentation of the patient, although the newer techniques for
measuring unbound digoxin could also be helpful (BANNER et al. 1992). It is
interesting to note that unconventional medicines could also participate in
drug-test interactions. This is important to remember when unusual results are
obtained since such medicines often do not appear on the patient's drug list.
Such an interference was noted with kyushin - a Chinese medicine that is very
popular in Japan, where it is obtainable without prescription. This medicine
was responsible for digoxin levels of more than 2.5 ng/ml in a patient who was
on a daily dose of 0.25 mg digoxin and did not exhibit any signs of digoxin
toxicity. The main components of the drug have chemical structures similar to
those of digoxin and digitalis-like cardiotonic actions. One tablet of kyushin
had a digoxin-like immunoreactivity equivalent to 1.9,ug (TDX analyzer,
Abbot Laboratories), 1.5,ug (Du Pont Aca V analyzer) and 72,ug digoxin
(Enzymun-Test, Boehringer Mannheim). The different equivalencies are
attributed to differences in cross-reactivity of the antibody used in the
immunoassays.
Two healthy volunteers took a typical dose of kyushin - two tablets three
times a day, and digoxin-like immunoreactivity reached almost OAng/ml in
half a day. The authors concluded that digoxin serum levels should be
interpreted very carefully in patients taking Chinese medicine (FUSHIMI et al.
1989).
studied is the one which is ten times the highest therapeutic concentration
reported in the literature or, if unknown, the concentration calculated from
ten times the therapeutic daily dose diluted in 5 or 151, depending on the
volume of distribution of the drug. All the drug and control samples should be
tested in duplicate.
If an interference is established, we follow with studies to measure the
magnitude of the analytical interference. At least five different concentrations
are used with two of them in the therapeutic range. A dose-effect curve is
obtained and the slope calculated.
Further steps used in the various studies for the detection and evaluation
of a drug-laboratory method interaction are the repetition of the study using
many different assays after establishing the basic difference in the results
obtained by them, that is when no interfering substance is present. The studies
should then be carried out on volunteers' or patients' biological fluids after
the consumption of the potential interfering drug in various dosages or
concentrations.
D. Conclusions
In the present chapter, an attempt has been made to shed light on a very
important phenomenon, the effect of drugs on laboratory tests in clinical
practice. Laboratory tests are an indispensable tool for appropriate diagnosis
and therapy, and we need to be able to rely upon them. We must strive to
make them highly specific but, when this is not possible, the alternative is to
become familiar with the list of drug-test interferences. Knowledge of the
mechanisms of the interferences will assist us greatly in remembering and in
understanding them.
When we come across a suspected drug-test interference, there are certain
practical steps that we should follow. This is well presented in the suggested
algorithm by TROUB (1992). When the laboratory result is inconsistent with
other laboratory results or the clinical picture, the possibility of transcription
error should be ruled out and, if no such error is found, the test should be
repeated (with the new and original specimens). If the doubtful results are
consistent, a pharmacological or dietary effect should be ruled out. The litera-
ture should then be consulted for past documentation of possible analytical
interference. If this is found, either the offending drug should be discontinued
and the test repeated, or, if this is not possible, a different method of analysis
should be employed.
Whenever we come across a newly suspected drug-laboratory test inter-
ference, it should be made known to the medical community, so that others
will be able to perform further studies to evaluate the nature of the interfer-
ence and suggest ways to eliminate it. The documentation provided by such
studies will assist in minimizing possible errors in diagnosis, as well as in
treatment.
322 S. YOSSELSON-SUPERSTINE
References
Al Hujaj M, Schonthal H (1971) Hyperuricemia and levodopa. N Engl J Med 285:859-
860
AI-Hujaj M, Schonthal H (1972) Gout and levodopa. N Engl J Med 286:376
Alper C, Seitchik J (1957) Comparison of the Archibald-Ken and Stransky colorimetric
procedure and the Praetorius enzymatic procedure for the determination of uric
acid. Clin Chern 3:95-101
Altman DG, Bland JM (1974) Diagnostic tests 1: Sensitivity and specificity. Br Med J
308:1552
Anon (1980) Diuretics, hyperuricemia and tienilic acid. Lancet 11:681-682
Anon (1982) Proteins in urine. In: Modern urine chemistry, Ames Division, Miles
Laboratories, Elkhart, Indiana
Anon (1993) Drug-induced discoloration of feces and urine. In: Knoben E, Anderson
PO (eds) Handbook of clinical drug data. Drug Intelligence Publications,
Hamilton, Illinois, p 22
Anon (1994) Digoxin-quinidine. In: Tatro DS, Olin BR, Hebel SK (eds) Drug interac-
tion facts. Facts and comparisons, St. Louis, p 290
Banner JW, Bach P, Burke B, Freestone S, Gooch WM (1992) Influence of assay
methods on serum concentrations of digoxin during Fab fragment treatment. Clin
ToxicoI30:259-267
Bjerve KS, Egens J, Lampinen LM, Masson P (1988) Evaluation of several creatinine
methods in search of a suitable secondary reference method: report from the
subcommittee on reference method for creatinine. Nordic Society for Clinical
Chemistry. Scand J Clin Lab Invest 48:365-373
Bottorff MB, Stewart CF (1986) Analytical techniques and quality control. In: Taylor
WJ, Diers Caviness MH (eds) A textbook for the clinical application of therapeu-
tic drug monitoring. Abbott Laboratories, Texas, pp 51-57
Bradley M, Schuman GB, Ward PCJ (1979) Examination of urine. In: Henry JB (ed)
Clinical diagnosis and management by laboratory methods. WB Saunders Com-
pany, Philadelphia, pp 559-634
Brass EP (1984) Effects of hypertensive drugs on endocrine function. Drugs 27:447-
458
Bruns DE (1988) Lactulose interferes in the alkaline picrate assay for creatinine. Clin
Chern 34:2592-2593
Caraway WT (1963) Uric acid. Stand Meth Clin Chern 4:239-247
Caraway WT (1969) Non-urate chromagens in body fluids. Clin Chern 24:54-57
Caraway WT, Marable H (1966) Comparison of carbonate and uricase-carbonate
methods for the determination of uric acid in serum. Clin Chern 12:18-24
Cawein MJ, Hewins J (1969) False rise in serum uric acid after L-dopa. N Engl J Med
281:1489-1490
Couttie K, Earle J, Loakley J (1987) Evaluation of single-slide creatinine method on
the Kodak Ektachem 700 shows positive interference from lidocaine metabolites.
Clin Chern 33:1674
Coyle JD, Mackichan JJ, Boudoulas H, Lima JJ (1987) Reversed-phase liquid chroma-
tography method for measurement of procainamide and three metabolites in
serum and urine: percent of dose excreted as deethyl metabolites. J Pharm Sci
76:402-405
Datta P, Graham GA, Schoen I (1986) Interference by IgG paraproteins in the Jaffe
method for creatinine determination. Am J Clin Pathol 85:463-468
Ducharme MP, Smythe M, Strohs G (1993) Drug induced alterations in serum creati-
nine concentrations. Ann Pharmacother 27:622-633
Dutcher JS, Strong JM (1977) Determination of plasma procainamide and N-
acetylprocainamide concentration by high pressure liquid chromatography. Clin
Chern 23:1318-1320
Ejvinsson G (1978) Effect of quinidine on plasma concentrations of digoxin. Br Med J
1:279-280
Drugs Causing Interference with Laboratory Tests 323
Engle JP, Donnelly AJ, Lewis RK (1988) Fecal occult blood tests: potential drug
interactions. Consult Pharm 3:356-358
Folin 0, Denis W (1912) A new colorimetric method for the determination of uric acid
in blood. J BioI Chern 13:469-475
Fushimi R, Tachi J, Amino N, Miyai K (1989) Chinese medicine interfering with
digoxin immunoassays. Lancet 1:339
Gaddis GM, Gaddis ML (1990) Introduction to biostatistics: part 3. Sensitivity,
specificity, predictive value, and hypothesis testing. Ann Emerg Med 19:591-
597
Galteau MM, Siest G (1984) Drug effects in clinical chemistry. Part 2. Guidelines for
evaluation of analytical interference. J Clin Chern Clin Biochem 22:275-279
Gannon RH, Levy RM (1984) Interference of third-generation cephalosporins with
theophylline assay by high-performance liquid chromatography. Am J Hosp
Pharm 41:1185-1186
Gannon RH, Phillips LR (1982) Metronidazole interference with procainamide HPLC
assay. Am J Hosp Pharm 39:1966-1967
Gault MH, Longerich L, Dawe M, Vasder SC (1985) Combined liquid chromatogra-
phy/radioimmunoassay with improved specificity for serum digoxin. Clin Chern
31:1272-1277
Gibaldi M, Grundhofer B, Levy G (1974) Effect of antacids on pH of urine. Clin
Pharmacol Ther 16:520-525
Gibb I, Adams PC, Pamham AJ, Jenning K (1983) Plasma digoxin: assay anomalies in
Fab-treated patients. Br J Clin Pharmacol 16:445-447
Gosney K, Adachi-Kirkland J, Schiller HS (1987) Evaluation of lidocaine interference
in the Kodak Ektachem 700 Analyzer single-slide method for creatinine. Clin
Chern 33:2311
Grayzel AL, Liddle L, Seegmiller JE (1961) Diagnostic significance of hyperuricemia
in arthritis. N Engl J Med 265:763-768
Green AJE, Halloran SP, Mould GP (1990) Interference by newer cephalosporins in
current methods for measuring creatinine. Clin Chern 36:2139-2140
Grotsch H, Hajdu P (1987) Interference by the new antibiotic cefpirome and other
cephalosporins in clinical laboratory tests, with special regard to the "Jaffe" reac-
tions. J Clin Chern Clin Biochem 25:49-52
Gyure WL (1977) Comparison of several methods for semiquantitative determination
of urinary protein. Clin Chern 23:876-879
Hansten PD (1992) Drug interactions. In: Koda-Kimble MA, Young LY (eds) Applied
therapeutics, the clinical use of drugs. Applied Therapeutics, Vancouver, Wash-
ington, pp 2-1-2-12
Haeckel R (1981) Assay of creatinine in serum with use of Fuller's earth to remove
interferents. Clin Chern 27:179-183
Henry RJ, Sobel C, Kim J (1957) A modified carbonate-phosphotungstate method for
the determination of uric acid and comparison with the spectrophotometric
uricase method. Am J Clin PathoI28:152-160
Herrington D, Drusano GL, Smalls U, Standiford HC (1984) False elevation in serum
creatinine levels. JAMA 252:2962
Hickman PE, Mather P (1988) Variation among instruments in interference by cepha-
losporin in the Jaffe reaction for creatinine. Clin Chern 34:215-216
Honda H, Gindin RA (1972) Gout while receiving levodopa for parkinsonism. JAMA
219:55-57
Huffman DH (1974) The effect of spinonolactone and canrenone on digoxin radioim-
munoassay. Res Commun Chern Pathol Pharmacol 9:787
Insel P A (1990) Analgesic-antipyretics and antiinflammatory agents: drugs employed
in the treatment of rheumatoid arthritis and gout. In: Gilman AG, RaIl TW, Nies
AS, Taylor P (eds) Goodman and Gilman's The pharmacological basis of thera-
peutics. Pergamon Press, New York, pp 638-681
Jaffe M (1886) Uber den Niederschlag we1chen Pikrinsaeure in normalen Ham erzeugt
und tiber eine neue Reaktion des Kreatinins. Z Physiol Chern 10:391-400
324 S. YOSSELSON-SUPERSTINE
A. Introduction
Medicines derived from plants formed the majority of the earlier materia
medica because chemically synthesised compounds were then not available.
Many of these herbs have stood the test of time and critical clinical assessment
and have found their way into the pharmacopoeias of orthodox medicines
sometimes as the isolated and chemically standardised active ingredient. Such
drugs as cocaine, colchicine, coumarin anticoagulants, digoxin, ephedrine,
morphine, quinine and quinidine, reserpine, tubocurarine, sennosides, and the
ergot and vinca alkaloids entered orthodox medicinal use by this route.
There are many types of herbal remedies, ranging from self-made teas
prepared from self-collected herbs to officially registered drug products which
have passed through the same rigid registration procedure as synthetic medi-
cines (see Table 1).
It is rather difficult to classify a particular herb in its most appropriate
category. The same botanical product which is a registered medicine in one
country may be a dietary supplement or recreational herb in the next, and in
one country the same herb may be available as an official medicine, as a
health food preparation and as a raw ingredient (DE SMET 1993a). Yet it is
important to keep these different categories in mind, when the risks of herbal
remedies are discussed, because the nature and magnitude of these risks can
vary considerably with the specific type of product. For instance, a consumer
of a high-quality registered herbal medicine should not have to worry about
the correct identity of the ingredients, whereas this should be a primary
concern for an individual who goes out into the field to collect his own herbal
materials.
The majority of preparations used in herbal or non-orthodox medicines
are a mixture of herbal ingredients. Herbs having diverse actions may be
incorporated into one concoction. Also herbal products are, in some circum-
stances, a mixture of herbal remedies with other ingredients of non-herbal
origin (e.g., arsenic or lead) or undeclared Western drugs (e.g., prednisolone,
non-steroidal anti-inflammatory/antirheumatic agents and paracetamol) as
found recently by KARUNANITHY and SUMITA (1991) in traditional Chinese
antirheumatic medicines. Readers are also referred to the reviews of D'ARCY
(1991,1993) and to a comprehensive review by DE SMET (1992a) on adultera-
328 P.A.G.M. DE SMET and P.F. D'ARCY
tion and contamination of herbal products with toxic metals and synthetic drug
substances and other drugs used in non-orthodox medicine.
It must be appreciated that the quality control exerted over most herbal
preparations and other non-orthodox remedies which are not registered as
medicines is often poor and more likely to be non-existent and that most
preparations are not standardised for potency in biological test systems. As a
consequence their potency may vary considerably from sample to sample.
Some of the interactions indicated in this review are based on a firm
pharmacological basis but their clinical relevance still needs to be established.
Others have been presented in anecdotal reports with lack of data essential to
establish a firm cause-effect relationship. In conformity with the objective of
this volume, mechanisms of the interaction are given where this is possible;
that it is not always possible is acknowledged since often the available data do
not permit even an assumption of the cause.
This approach in the context of this chapter is necessary if warnings are to
be given about likely interactions since the sparse reports of drug interactions
between non-orthodox products and Western medicines neither confirms their
safety in use nor indeed suggests that the incidence of such interactions is low.
The simple fact is that most interactions of this type will not be recognised as
such by the self-medicated patient and will not be reported to an orthodox
medical practitioner and therefore will not appear in the medical or pharma-
ceuticalliterature.
When such original reports of reactions and interactions, as are available,
are carefully analysed, it becomes obvious that many of the cases where herbal
products have been associated with actual human poisoning were not in fact
caused by the herbs alleged to be in the product, but resulted from substitution
or contamination of the declared ingredients, intentionally or by accident, with
a more toxic botanical, a poisonous metal, or a potent non-herbal drug sub-
stance (DE SMET 1992a).
Although herbal medicines are by far the largest component of non-
orthodox remedies they do not have exclusive claims; it must be made clear
that there are various types of other alternative treatments ranging from
preparations of animal origin, minerals, vitamins and amino acids. Many of
Drug Interactions with Herbal and Other Non-orthodox Remedies 329
B. Animal Agents
There are few parts of animals that have not been used at one time or another
for their medicinal properties. For example, crude thyroid hormones may
occur as ingredients of slimming preparations and/or adulterants of herbal
products (DE SMET 1992a).
I. Fish Oil
Fish oil can upset coagulation control by increasing the bleeding time and
reducing platelet aggregation. It is difficult therefore to exclude the possibility
of untoward consequences in stabilised anticoagulated patients if this oil is
taken concomitantly with anticoagulant medication (DE SMET 1989).
c. Amino Acids
I. L- Tryptophan
D. Vitamins
Vitamin and mineral supplements are a common combination and are readily
obtainable as over-the-counter medicines; they may be regarded by consumers
as food supplements rather than medication and the possibility of their inter-
action with prescribed medicines may not be appreciated. Many drugs are
known to evoke vitamin deficiency states, for example isoniazid, hydralazine
and penicillamine are antagonistic to vitamin B6; anticonvulsants cause folate
and vitamin D deficiency and have been implicated in osteomalacia (DENT et
al. 1970); and vitamin B12 deficiency may occur after prolonged treatment with
the antidiabetic metformin (CALLAGHAN et al. 1980; ADAMS et al. 1983).
They can influence the effects of various orthodox drugs and sometimes
this may be used intentionally as an advantage (see SOKOL et al. 1991), but
more often unwanted effects occur. For example health foods and food
supplements containing appreciable quantities of vitamin K can reduce the
effect of oral anticoagulants (HEALD and POLLER 1974; STOCKLEY 1991). Cases
of impaired anticoagulation have been recorded after concomitant intake of
"Gon" (a non-orthodox remedy containing vitamin K4), enteral nutrition, and
excessive amounts of green vegetables (UDALL and KROCK 1968; HEALD and
POLLER 1974; HOGAN 1983; KEMPIN 1983; WALKER 1984; WATSON et al. 1984).
Vitamin B6 can nullify the beneficial effects of levodopa in parkinsonism
(COTZIAS 1969; CALNE and SANDLER 1970). A further example is that folic acid
may occasionally decrease serum phenytoin to a clinically significant degree
(HANSTEN et al. 1993). Vitamins may also influence the activity of other non-
orthodox remedies, for example the absorption of selenium from sodium
selenite can be drastically reduced by megadoses of vitamin C (ROBINSON et al.
1985), and the use of large doses of vitamin C (up to 5 g/day), taken prophylac-
tically against the common cold, may alter urinary pH sufficiently to influence
Drug Interactions with Herbal and Other Non-orthodox Remedies 331
the renal elimination and therefore the clinical response of alkaline drugs
(MEDICAL LETTER 1971).
The amount of vitamin consumed is dependent on the specific product and
its specific dose recommendations. While many multivitamin preparations
provide daily amounts below or just above their Recommended Daily Allow-
ance (RDA) values, there are also products which provide megadoses of one
or more vitamins, This latter category entails a risk of toxic effects (EVANS and
LACEY 1986; BROWN and GREENWOOD 1987; HELSING 1992) as well as drug
interactions. In general, however, with the majority of multivitamin/mineral
supplements, it is doubtful whether the amount of vitamin contained in the
product plays any great part in interactions with orthodox drugs.
E. Minerals
It is normally the mineral salt component of the combination as well as the
mineral content of antacids and some laxatives that is responsible for the
majority of interactions. For example, the absorption of tetracyclines is re-
duced by the formation of insoluble chelates in the gut in the presence of milky
and other foods containing bivalent and trivalent metal ions (Ca, Mg, AI, Fe)
(KUNIN and FINLAND 1961; BRAYBROOKS et a1. 1975; CHIN and LACH 1975), and
serum levels of oral tetracyclines may be decreased by more than 50% if they
are taken with ferrous sulphate or milk (NEUVONEN 1976).
Iron supplements are among the most frequently prescribed drugs (LA
PlANA SIMONSEN 1989) and are taken in over-the-counter remedies by many
other people. CAMPBELL and HASINOFF (1991) have listed some 19 orally
administered drugs, including antimicrobials, captopril, levodopa, salicylic
acid and thyroxine, which have functional groups in a configuration that will
bind iron to form an iron-drug complex. The formation of a stable iron-drug
complex reduces the extent of drug absorption but does not appear to reduce
the rate of drug absorption. Iron can also catalyse oxidation and reduction
interactions. Zinc also can produce various interactions with orthodox drugs
(BARRY et a1. 1991).
Materials with a high calcium content may reduce the absorption of the
bisphosphonates. Vitamins with mineral supplements such as iron, calcium
supplements, laxatives containing magnesium, or antacids containing calcium
or aluminium all dramatically reduce the biological availability of the
bisphosphonates (FLEISCH 1987; FELS et a1. 1989).
F. Dietary Fads
Dietary fads may lead to an excessive intake of certain nutrients, which in turn
could result in interference with orthodox medication. Certain diets, including
vegetable-rich weight-reducing diets, are rich in vitamin K and if taken in
sufficient amounts will interfere with warfarin treatment (HEALD and POLLER
332 P.A.G.M. DE SMET and P.F. D'ARCY
1974; HOGAN 1983; PATRIACA et al. 1983; WALKER 1984; WATSON et al. 1984).
For example, a patient maintained on warfarin 8mg daily required an increase
in dosage to 13 mg/day when placed on enteral nutrition rich in vitamin K; the
warfarin dosage returned to 8mg/day when the food supplement was stopped.
Problems have also been experienced with anticoagulated patients consuming
large amounts of broccoli (KEMPIN 1983; STOCKLEY 1984).
Beside the risk of pharmacodynamic problems, there is also the possibility
that the pharmacokinetic fate of orthodox drugs could be affected by a dietary
change. For instance, dietary fibres may affect absorption (see below),
whereas certain other foodstuffs may exert an influence on drug biotransfor-
mations in humans (ALVARES 1984; ANDERSON 1988). Also certain foods and
food supplements can affect the urinary excretion of drugs. A balanced diet
with adequate protein provides an acid urine, whereas a low-protein diet or a
strict vegetarian diet may give an alkaline urine. This opens up the possibility
that diet-induced changes in urinary pH affect the rates of excretion of weakly
acidic or weakly basic drugs (WESLEY-HADZIJA 1971; PIERPAOLI 1972). Acid
urine favours the ionisation of alkaline drugs (and vice versa), so reabsorption
is reduced and renal excretion is increased. Acid fruit juices and squashes may
therefore reduce the efficacy of the antimalarials quinine and chloroquine
(P.F. D'ARCY, personal observation).
Grapefruit juice, but not orange juice, greatly augments the bioavailability
of the antihypertensive, calcium-antagonists felodipine, nifedipine and
nitrendipine (BAILY et al. 1989, 1991; SOONS et al. 1991; EDGAR et al. 1992);
similar results are apparent on the clearance of caffeine (FUHR et al. 1993).
These affects are due to the inhibition of cytochrome P450 (isoforms CYP1A2
and CYP3A4) enzymes by several flavonoid glycosides present in grapefruit
juice, of which the bitter principle naringin (4',5,7-trihydroxyflavanone 7-
rhamno-glucoside) is the most abundant (FUHR et al. 1993).
Interestingly, grapefruit juice has also recently been found to inhibit the 7-
hydroxylation of coumarin, as measured by its effect on urinary excretion of 7-
hydroxycoumarin in healthy volunteers (MERKEL et al. 1994). This implies that
grapefruit juice may interfere with the pharmacokinetics of coumarin-yielding
medicinal herbs, such as Melilotus officinalis (sweet clover), Asperula odorata
(sweet woodruff), Dipteryx odorata (tonka bean) and Anthoxanthum
odoratum (sweet vernal grass) (HOPPE 1975; LEWIS and ELVIN-LEWIS 1977;
TEUSCHER and LINDEQUIST 1980). A comprehensive review on the significance
of interactions between grapefruit juice and drugs was recently presented by
BAILEY et al. (1994).
G. Herbal Drugs
This category includes crude herbal remedies as well as plant-derived drugs;
discussion will be divided into the following categories:
Drug Interactions with Herbal and Other Non-orthodox Remedies 333
1. I>ietary Fibres
BROWN et al. (1977) showed that digoxin bioavailability is decreased by almost
20% when given with a high-fibre meal. In vitro studies by FLOYD et al. (1977)
supported this and indicated that up to 45 % of the digoxin may be sequestered
in or bound to the bran. Later work by REISSELL and MANNINEN (1982)
showed that the effect of fibre was small but that an interactive effect on
absorption of digoxin might occur if fibre and digoxin were ingested simulta-
neously.
ROSSANDER (1987) suggested, on the basis of in vitro studies, that dietary
fibres might interfere with the bioavailability of iron in the diet. Confirmatory
work in healthy volunteers showed that bran had a marked inhibitory effect on
the absorption of dietary iron but not cellulose or pectins. ROE et al. (1988)
showed that dietary fibre in combined wheat bran and psyllium biscuits or
biscuits containing psyllium alone reduced the absorption of riboflavin by
6.4% and 5.7%, respectively. No effect of the wheat bran supplement alone
was detected.
The possibility that drug interactions with dietary fibre or other bulk-
forming herbal drugs may interfere with the gastrointestinal absorption of
orthodox drugs has become of more concern and there have been a number of
pivotal publications. For example, PERLMAN (1990) has reported a case which
raises the possibility of an interaction between lithium salts and ispaghula husk
(psyllium hydrophilic mucilloid), a bulk-forming laxative.
The patient had a long history of psychiatric problems and was treated
with lithium carbonate and then lithium citrate. She also received ispaghula
husk (one teaspoonful in water twice daily). Despite increasing her lithium
dosage, her blood lithium concentrations fell to below acceptable levels.
Ispaghula husk was discontinued and her blood lithium concentrations in-
creased.
RICHTER et al. (1991) have reported that concomitant fibre intake (pectin
15 mg/day or oat bran 50-100 g/day) may decrease the absorption of the lipid-
334 P.A.G.M. DE SMET and P.F. D'ARCY
2. Gnar Gnm
TODD et al. (1990) have reviewed the pharmacological properties of guar gum.
Administration of up to 30 g/day guar gum to diabetic patients does not seem
to alter mineral or electrolyte balance during long-term therapy (BEHALL et al.
1989; McivOR et al. 1985; UUSITUPA et al. 1989), although plasma levels of fat-
soluble vitamins (A and E) have tended to decrease during extended treat-
ment, but not to a clinically important extent (UUSITUPA et al. 1989).
Because of its effect on prolonging gastric retention and because drugs
will diffuse more slowly out of viscous matrices than from solutions, guar gum
may affect the absorption of certain concomitantly administered drugs. Often
only the rate of absorption is affected [e.g., bumetanide, digoxin, paracetamol
(acetaminophen)] and such interactions are of minor clinical significance.
However, the absorption of certain other drugs (e.g., metformin (GIN et al.
1989), phenoxymethylpenicillin (HUUPPONEN et al. 1985) and some formula-
tions of glibenclamide (NEUGEBAUER et al. 1983) may be reduced by a clinically
significant degree.
Guar gum is a component of many slimming preparations and it might be
advisable that women taking oral contraceptives take additional contraceptive
precautions whilst taking guar gum (ANONYMOUS 1987). The same warning
could be extended to women taking low-dose oestrogen, combined oral con-
traceptives which are on on any slimming diet that contains bulk-forming
agents. Theoretically, they could be at risk of contraceptive failure if the
absorption of the contraceptive is compromised. However, we know of no
clinical study that has assessed the clinical importance of this theoretical
notion, nor any report that has linked slimming diets with contraceptive
failure.
3. Tannins
From the 1960s to the present time, it has been variously suggested and refuted
that among the botanical constituents that might also interfere with the ab-
sorption of some orthodox drugs are tannins in tea or coffee (CARRERA et al.
1973) and phytates (DAVIES 1982). For example, CARRERA et al. (1973) showed
in rat studies that the absorption of vitamin B12 was reduced proportionately to
the oral dose of tannic acid administered. This interaction, it was claimed, was
Drug Interactions with Herbal and Other Non-orthodox Remedies 335
1. Cola Nut
An extensive study by FRASER et al. (1976) of factors affecting antipyrine
metabolism in West African villagers showed that cola nut consumption inhib-
ited antipyrine metabolism and prolonged the antipyrine half-life by 3.Sh. It
was suggested that unidentified constituents of cola nuts competed with
antipyrine for oxidation by the microsomal enzyme system. There were three
other predictors of oxidative capacity: sex, haemoglobin in women and height
in men.
VESELL et al. (1979) failed to show any effect of cola nuts, chewed for
either 14 or 28 consecutive days, on antipyrine disposition in Caucasian males.
Genetic factors may therefore be of importance and although many questions
remain unanswered, the results of the antipyrine studies are interesting
enough to suggest that this type of work be reopened since further research in
this direction and particularly with other herbal remedies would seem to be
warranted.
2. Eucalyptus Species
A number of studies have demonstrated that eucalyptus leaves, the oil and the
major active principle, eucalyptol, can all induce microsomal enzyme activity
in both in vitro and in vivo tests (JORI et al. 1969; SEAWRIGHT et al. 1972).
However, there have not been any recorded interactions between eucalyptus
and orthodox drugs at the clinical level.
3. Grapefruit Juice
There is evidence that grapefruit juice augments the bioavailability of the
antihypertensives felodipine, nifedipine and nitrendipine, and the clearance of
caffeine. These interactions have already been mentioned previously under
"Dietary Fads".
5. Kampo Medicines
Oriental Kampo medicines often contain glycyrrhizin and these have been
reported to influence prednisolone pharmacokinetics. HOMMA et al. (1992a)
Drug Interactions with Herbal and Other Non-orthodox Remedies 337
showed that three Kampo remedies, which also contained Saiko (Bupleuri
radix), and were commonly co-administered with prednisolone in the treat-
ment of asthma, nephrotic syndrome and collagen diseases, had a variable and
different effect on prednisolone pharmacokinetics in healthy sUbjects.
Only one of the three remedies had a steroid-sparing effect due to de-
creased l1,B-hydroxysteroid dehydrogenase (ll-beta-HSD) activity. The other
two either increased or did not change the activity of this enzyme. Interest-
ingly, the authors attributed the enzyme inhibitory effect not to glycyrrhizin,
which was present in the three Kampos, but to magnolol, which was only
contained in the Kampo with a positive effect on prednisolone metabolism.
Follow-up work by HOMMA et al. (1994) was aimed at confirming the
inhibitor of prednisolone metabolism contained in Saiboku-To. In in vitro
experiments they studied the effects of 10 herbal constituents on ll-beta-HSD
and showed that five herbal extracts had inhibitory activity: Glycyrrhiza glabra
> Perillae frutescens > Zizyphus vulgaris> Magnolia officinalis > Scuttellaria
baicalensis. Seven chemical constituents which were identified as the major
urinary products of Saiboku-To in humans were studied; magnolol derived
from M. officina lis showed the most potent inhibition of the enzyme and
although this was less than that of glycyrrhizin, the non-competitive inhibition
mechanism was different from a known competitive mechanism. These results
suggested that magnolol might contribute to the inhibitory effects of Saiboku-
To on prednisolone metabolism through inhibition of ll-beta-HSD.
Studies on ofloxacin in healthy volunteers by HASEGAWA et al. (1994)
found no significant effects of the Kampo medicines Sho-saiko-To (TJ-9),
Rikkunshi-To (TJ-43) and Sairei-To (TJ-1l4) on any estimated bioavailability
parameter.
6. Liquorice
Liquorice, the dried rhizome and roots of Glycyrrhiza glabra, has long been a
popular and traditional ingredient of both herbal and orthodox medicines. It is
also found in confectionary, soft drinks, chewing tobacco and chewing gum.
There is evidence to suggest that liquorice may interfere with the pharmacoki-
netics of orthodox medicines. For example, CHEN et al. (1990) showed that an
intravenous infusion of the active principle, glycyrrhizin (GL), increased the
plasma concentrations of prednisolone and influenced the pharmacokinetics
of prednisolone in man. Interestingly, the main component in plasma is differ-
ent when GL is administered by different routes. After intravenous adminis-
tration the main component in plasma is glycyrrhizin (KATO et al. 1984; CHEN
et al. 1990), while after oral dosage the component is glycyrrhetinic acid (GA),
a metabolic derivative of GL (NAKADA et al. 1986; SHIMADA et al. 1989).
In vitro, the inhibiting effect of GA on the metabolism of corticosteroids
by 5a-,5,B-reductase and ll,B-dehydrogenase is stronger than that of GL. It is
not surprising therefore that in later studies CHEN et al. (1991) showed that
oral administration of GL also modified the pharmacokinetics of both total
338 P.A.G.M. DE SMET and P.F. D' ARCY
and free prednisolone. The area under the curve (AUC) of prednisolone was
significantly increased, the total plasma clearance was significantly reduced,
and the mean residence time was significantly prolonged. However, the vol-
ume of distribution showed no evident change.
These results suggested that the oral administration of GL increases
the plasma prednisolone concentrations and influences its pharmacokinetics
by inhibiting its metabolism, but not affecting its distribution. It has been
suggested that this combination would be advantageous in the treatment
of rheumatoid conditions. The basis of the interaction has been suggested
as inhibition of the metabolism of prednisolone by microsomal enzymes;
inhibition of urine clearance and interference with plasma protein binding
were both discounted. There is direct evidence that GL and GA can inhibit
corticosteroid 5a- and 5p-reductase and IIp-dehydrogenase activities in
rat liver and kidney in vitro and in vivo (TAMURA et al. 1979; MONDER et al.
1989).
1. Caffeine-Containing Herbs
Certain antibacterial 4-quinolones and fluoroquinolones (ciprofloxacin,
enoxacin, pipemidic acid and temafloxacin) inhibit the hepatic metabolism of
caffeine, increase its elimination half-life and decrease its clearance (CARBO et
al. 1989; HARDER et al. 1989; HEALY et al. 1989; MAHR et al. 1992). Users of
caffeine-containing beverages and herbals should therefore be advised that
they have an increased risk of adverse effects (e.g., tremor, tachycardia, insom-
nia, CNS excitation) when they are taking such quinolones. The most impor-
tant herbal remedies which contain substantial amounts of caffeine are
derived from Cola, !lex and Paullinia species (DE SMET 1989).
2. Sparteine-Containing Herb
Sparteine is a quinolizidine alkaloid from Cytisus scoparius which was recently
found in a herbal slimming remedy on the United Kingdom market. Substan-
tial doses of this preparation in slow metabolisers could be expected to
be associated with many adverse reactions including circulatory collapse
(GALLOWAY et al. 1992). The antiarrhythmic agent quinidine is a potent inhibi-
tor of the oxidative metabolism of sparteine (SCHELLENS et al. 1991) and a
similar effect has been observed with haloperidol (GRAM et al. 1989) and with
moclobemide (GRAM et al. 1993).
In addition, CREWE et al. (1992) conducted in vitro experiments in which
several antidepressants (tricyclic or selective serotonin reuptake inhibitors)
Drug Interactions with Herbal and Other Non-orthodox Remedies 339
3. Teucrium chamaedrys
LOEPER et al. (1994) evaluated the molecular mechanism of the hepatotoxicity
which had been observed in numerous users of a French preparation contain-
ing Teucrium chamaedrys. A dose-dependent increase in serum alanine ami-
notransferase (ALT) activity in mice was observed after the intragastric
administration of the lyophilisate of a tea prepared from blooming aerial parts.
Hepatotoxicity could also be produced by intragastric administration of
0.125 g/kg of an enriched fraction, which contained the same level of furano
neo-clerodane diterpenoids as 1.25 g/kg of the lyophilisate. The increase
in serum ALT activity could be enhanced by inducers of the 3A family
(troleandomycin). Toxicity was also increased by pretreatment with phorone
(a depletor of hepatic glutathione), whereas it could be attenuated by inducers
of microsomal epoxide hydrolase (such as clofibrate). These findings suggest
that the hepatotoxicity of T. chamaedrys resides in one or more reactive
metabolites of its furanoditerpenoids and that orthodox drugs may influence
their formation.
Table 2. Herbal drugs with well-known constituents and their interactions. (DE SMET
1992b; D' ARCY 1993)
heavy betel nut chewing. The mechanism for this effect was suggested as
antagonism of the anticholinergic agent procyclidine, by the active alkaloid
ingredient of the betel, arecoline (DEAHL 1989).
TAYLOR et al. (1992) have included betel nut chewing among the factors
that dispose to asthma severity and unsatisfactory control by orthodox medi-
cines in Asians residing in the United Kingdom. It was suggested that
arecoline, or another alkaloid in betel nut, for example, guvacoline, may have
a cholinergic bronchoconstrictor effect.
5. Picrorhiza ku"oa
According to BEDI et al. (1989), the rhizomes of this plant species, which are
used in Ayurvedic medicine, might potentiate the photo chemotherapeutic
effects of methoxsalen in human patients with vitiligo.
1. Anthranoid Laxatives
An early German report suggests that quinidine serum levels might be re-
duced by the concurrent use of herbal anthranoid laxative preparations, such
342 P.A.G.M. DE SMET and P.F. D'ARCY
2. Berberine
Berberine is an alkaloid derived from the roots and bark of the plant Berberis
aristata (barberry bush); extracts of this plant have been used in antidiarrhoeal
medication in Ayurvedic medicine in India and in the traditional medicine of
China for the past 3000 years. A Burmese study on the clinical effects of
berberine in acute watery diarrhoea, namely the reputed antisecretory and
vibriostatic effect, showed that berberine alone did not benefit the duration of
diarrhoea, frequency of stools and fluid requirements for rehydration, nor did
it produce a noticeable antisecretory effect. Clinically, patients with cholera
given tetracycline plus berberine were more ill, suffered longer from diarrhoea
and required larger volumes of intravenous fluid than did those given tetracy-
cline alone (KHIN-MAUNG-U et al. 1985).
4. Piperine
There are several studies to show that piperine, a major alkaloid of Piper
longum and P. nigrum, both of which occur in Ayurvedic formulations, can
enhance the bioavailability of orthodox drugs such as phenytoin, propranolol,
rifampicin, sulphadiazine, tetracycline and theophylline. Among the suggested
mechanisms are promotion of gastrointestinal absorption, inhibition of drug
metabolism and a combination of these two (ATAL et al. 1981, 1985; BANo et
al. 1987, 1991; BHAT and CHANDRASEKHARA 1987; JOHRI and ZUTSHI 1992).
5. "Shankhapusphi"
This is an Ayurvedic non-alcoholic syrup which is prepared from six herbs:
Centella asiatica, Convolvulus pluricaulis, Nardostachys jatamansi, Nepeta
elliptica, Nepeta hindostana and Onosma bracteatum. DANDEKAR et al. (1992)
have reported two epileptic patients taking phenytoin who experienced an
unexpected loss of seizure control and a reduction in plasma phenytoin levels
when they took this herbal preparation. A follow-up study in rats to investi-
gate this possible interaction showed that multidose co-administration (but
not single dose) reduced not only the antiepileptic activity of phenytoin but
also lowered plasma phenytoin levels. Shankhapusphi itself showed significant
Drug Interactions with Herbal and Other Non-orthodox Remedies 343
6. Yohimbine
Yohimbine is a toxic alkaloid from Pausinystalia yohimbe. Preparations pro-
viding pharmacologically relevant doses of yohimbine can occasionally be
encountered on the Western health food market, even though this is a toxic
alkaloid which is not sufficiently safe to be freely available for uncontrolled
use (DE SMET and SMEETS 1994). When given to healthy volunteers, 15-20mg
p.o. yohimbine is usually needed to increase blood pressure and to induce
anxiety, but in patients on tricyclic antidepressants hypertension may already
occur at 4mg t.i.d. (LACOMBLEZ et al. 1989). The toxicity of yohimbine can also
be enhanced by other drugs, such as chlorpromazine, while it is attenuated by
amobarbital or reserpine (INGRAM 1962).
Remarkably, yohimbine may be protrayed in off-label advertising of
yohimbe health food preparations as a peripheral vasodilator, which can po-
tentiate other blood pressure-lowering agents. In reality, however, the a-
adrenoreceptor antagonist properties of the alkaloid will reverse the effects of
clonidine and similar antihypertensives (DE SMET and SMEETS 1994).
H. Comment
The use of natural (herbal) and other non-orthodox medicines is a persistent
aspect of present-day health care and Europeans alone are thought to spend
the equivalent of US $500-600 million/year on natural remedies and food
supplements. Many consumers believe that naturalness is a guarantee of harm-
lessness and have no qualms, when necessary, in taking their own prescribed
conventional medicine as well. In the United Kingdom and Netherlands
and elsewhere in Europe many immigrant races have their own traditional
medicine practices, which they frequently combine with orthodox medical
care. Generally too little is known about the consequences of such combina-
tions, although the clinical reports of interactions that infrequently appear in
the medical and pharmaceutical press suggest that many more interactions
may be occurring that are not realised as such and are not reported in the
literature.
It should be realised that the data presented in this review did not always
come from studies intended and designed to evaluate adverse interactions
between orthodox drugs and herbal drugs. Some data were merely obtained as
a "spin-off" from pharmacological studies, in which the most useful probe
happened to be a herbal constituent (sparteine, coumarin) or which were
Drug Interactions with Herbal and Other Non-orthodox Remedies 345
safety of the available products. This approach can only work, however, if it is
supplemented by active herbal pharmacovigilance (DE SMET 1993b), which
should look not only for unknown adverse reactions but also for new adverse
drug reactions (DE SMET 1995).
References
Adams JF, Clarke JS, Ireland JT, Kesson CM, Watson WS (1983) Malabsorption of
vitamin B12 and intrinsic factor secretion during biguanide therapy. Diabetologia
24:16-18
Alvares AP (1984) Environmental influences on drug biotransformations in humans.
World Rev Nutr Diet 43:45-59
Anderson KE (1988) Influences of diet and nutrition on clinical pharmacokinetics. Clin
Pharmacokinet 14:325-346
Anonymous (1987) Guar gum of help to diabetics. Drug Ther Bull 25:65-66
Aslam M, Stockley IH (1979) Interactions between curry ingredient (Karela) and drug
(chlorpropamide). Lancet 1:607
Atal CK, Zutshi U, Rao PG (1981) Scientific evidence on the role of Ayurvedic herbals
on bioavailability of drugs. J Ethnopharmacol 4:229-232
Atal CK, Dubey RK, Singh J (1985) Biochemical basis of enhanced drug bioavailability
by piperine: evidence that piperine is a potent inhibitor of drug metabolism.
J Pharmacol Exp Ther 232:258-262
Bailey DG, Spence JD, Edgar B, Bayliff CD, Arnold JMO (1989) Ethanol enhances
the hemodynamic effects of felodipine. Clin Invest Med 12:357-362
Bailey DG, Spence JD, Munoz C, Arnold JMO (1991) Interaction of citrus juices with
felodipine and nifedipine. Lancet 337:268-269
Bailey DG, Arnold JM, Spence JD (1994) Grapefruit juice and drugs. How significant
is the interaction? Clin Pharmacokinet 26:91-98
Bano G, Raina RK, Sharma DB (1986) Pharmacokinetics of carbamazepine in protein
energy malnutrition. Pharmacology 32:232-236
Bano G, Amla V, Raina RK, Zutshi U, Chopra CL (1987) The effect of piperine on
pharmacokinetics of phenytoin in healthy volunteers. Planta Med 53:568-569
Bano G, Raina RK, Zutshi U, BdiKL, Johri RK, Sharnma SC (1991) Effect of piperine
on bioavailability and pharmacokinetics of propranolol and theophylline in
healthy volunteers. Eur J Clin PharmacoI41:615-617
Barry MG, Macmathuna P, Younger K, Keeling PWN, Feely J (1991) Effect of zinc
supplementation on oxidative drug metabolism in patients with hepatic cirrhosis.
Br J Clin Pharmacol 31:488-491
Bedi KL, Zutshi U, Chopra CL, Amia V (1989) Picrorhiza kurroa, an Ayurvedic herb,
may potentiate photochemothereapy in vitiligo. J Ethnopharmacol 27:347-352
Behall KM, Scholfield DJ, McIvor ME, Van Duyn M, Leo TA et al. (1989) Effects of
guar gum on mineral balances in NIDDM adults. Diabetes Care 12:357-364
Bhat BG, Chandrasekhara N (1987) Interaction of piperine with rat liver microsomes.
Toxicology 44:91-98
Bordia AK, Joshi HK, Sanadhya YK, Bhu N (1977) Effect of essential oil of garlic on
serum fibrinolytic activity in patients with coronary artery disease. Ather-
oscelerosis 28:155-159
Bowen S, Taylor KM, Gibb IAMcL (1981) Effect of coffee and tea on blood levels and
efficacy of antipsychotic drugs. Lancet 1:1217-1218
Braybrooks MP, Barry BW, Abbs ET (1975) The effect of mucin on the bioavailability
of tetracycline from the gastrointestinal tract in vivo, in vitro correlation. J Pharm
Pharmacol 27:508-515
Brown DD, Juhl RP, Warner SL (1977) Decreased bioavailability of digoxin produced
by dietary fiber and cholestyramine. Am J Cardiol 39:297
Brown GR, Greenwood JK (1987) Megavitamin toxicity. Can Pharm J 120:80-87
Drug Interactions with Herbal and Other Non-orthodox Remedies 347
Callaghan TS, Hadden DR, Tomkin GH (1980) Megaloblastic anaemia due to vitamin
B12 absorption associated with long-term metformin treatment. Br Med J 280:
1214-1215
CaIne DB, Sandler M (1970) L-Dopa and Parkinsonism. Nature 226:21-24
Campbell NRC, Hasinoff BB (1991) Iron supplements: a common cause of drug
interactions. Br J Clin Pharmacol 31:251-255
Capobianco DJ, Brazis PW, Fox TP (1993) Proximal-muscle weakness induced by
herbs. N Engl J Med 329:1430
Carbo M, Segura J, De la Torre R, Badenas JM, Cami J (1989) Effect of quinolones on
caffeine disposition. Clin Pharmacol Ther 45:234-240
Carrera G, Mitjavila S, Derache R (1973) Effect de l'acide tannique sur l'absorption de
la vitamine B12 chez Ie rat. C R Acad Paris [D] 276:239-242
Chang H-M, But PPH (eds) (1987) Pharmacology and applications of Chinese materia
medica. World Scientific, Singapore
Chen M-F, Shimada F, Kato H, Yano S, Kanaoka M (1990) Effect of glycyrrhizin on
the pharmacokinetics of prednisolone following low dosage of prednisolone
hemisuccinate. Endocrinol Jpn 37:331-341
Chen M-F, Shimada F, Kato H, Yano S, Kanaoka M (1991) Effect of oral administra-
tion of glycyrrhizin on the pharmacokinetics of prednisolone. Endocrinol Jpn
38:167-175
Chin TF, Lach JL (1975) Drug diffusion and bioavailability: tetracycline metallic
chelation. Am J Hosp Pharm 32:625-629
Cotzias GC (1969) Metabolic modification of some neurological disorders. JAMA
210:1255-1262
Crewe HK, Lennard MS, Tucker GT, Woods FR, Haddock RE (1992) The effect of
selective serotonoin re-uptake inhibitors on cytochrome P4502D6 (CYP2D6) ac-
tivity in human liver microsomes. Br J Clin Pharmacol 34:262-265
Dandekar UP, Chandra RS, Dalvi SS, Joshi MV, Gokhale PC, Sharma MV, Shah PU,
Kshirsagar NA (1992) Analysis of a clinically important interaction between
phenytoin and Shankhapushpi, an Ayurvedic preparation. J Ethnopharmacol
35:285-288
D'Arcy PF (1991) Adverse reactions and interactions with herbal medicines. I. Ad-
verse reactions. Adverse Drug React Toxicol Rev 10:189-208
D' Arcy PF (1993) Adverse reactions and interactions with herbal medicines. II. Drug
interactions. Adverse Drug React Toxicol Rev 12:147-162
Davies NT (1982) Effects of phytic acid on mineral availability. In: Vahouny GV,
Krtichevshky D (eds) Dietary fiber in health and disease. Plenum, New York, pp
105-116
De Freitas B, Schwartz G (1979) Effects of caffeine in chronic psychiatric patients. Am
J Psychiatry 136:1337-1338
De Smet P AGM (1985) Ritual enemas and snuffs in the Americas. Foris, Dordrecht
(Latin America studies no 33, Centre de Estudios y Documentacion
Latinoamericanos)
De Smet PAGM (1989) Drugs used in non-orthodox medicine. In: Dukes MNG,
Beeley L (eds) Side effects of drugs annual 13. Elsevier, Amsterdam, pp 442-
473
De Smet PAGM (1991) Drugs used in non-orthodox medicine. In: Dukes NMG,
Aronson JK (eds) Side effects of drugs annual 15. Elsevier, Amsterdam, pp 514-
531
De Smet PAGM (1992a) Toxicological outlook on the quality assurance of herbal
remedies. In: De Smet PAGM, Keller K, Hansel R, Chandler RF (eds) Adverse
effects of herbal drugs, voll. Springer, Berlin Heidelberg New York, pp 1-72
De Smet PAGM (1992b) Drugs used in non-orthodox medicine. In: Dukes MNG (ed)
Side effects of drugs. Elsevier, Amsterdam, pp 1209-1232
De Smet PAGM (1993a) Legislatory outlook on the safety of herbal medicines. In: De
Smet PAGM, Hansel R, Kellere K, Chandler RF (eds) Adverse effects of herbal
drugs, vol 2. Springer, Berlin Heidelberg New York, pp 1-90
348 P.A.G.M. DE SMET and P.F. D'ARCY
Griffin JP, D'Arcy PF, Speirs CJ (1988) A manual of adverse drug interactions, 4th edn.
Wright, (Butterworths) London
Guckenbiehl W, Gilfrich HJ, Just H (1976) EinfluB von Laxantien und Metoclopramid
auf die Chinidin-Plasmakonzentration wahrend Langzseittherapie bei Patienten
mit HerzrythmusstOrungen. Med Welt 27:1273
Hanawa T, Hirama N, Kosoto H, Ja Hyun S, Ohwada S, Hasegawa R, Haranaka R,
Nakagawa S (1987) Effects of Saiko-zai on collagen metabolism in mice adminis-
tered with glucocorticoid. Nohon Univ J Med 29:197-205
Hansten PD, Horn JR, Koda-Kimble MA, Young L Y (eds) (1993) Anticonvulsant
drug interactions - folic acid (Folvite). Drug Interact Updates Q 13:355-356
Harder S, Fuhr U, Staib AH, Wolff T (1989) Ciprofloxacin-caffeine: a drug interaction
established using in vivo and in vitro investigations. Am J Med 87:89S-91S
Hasegawa T, Yamaki K, Nadai M, Muraoka I, Wang L, Takagi K, Nabeshima T (1994)
Lack of effect of Chinese medicines on bioavailability of ofloxacin in healthy
volunteers. Int J Clin Pharmacol Ther 32:57-61
Heald GE, Poller L (1974) Anticoagulants and treatment for chilblains. Br Med J
2:455
Healy DP, Polk RE, Kanawati L, Rock DT, Mooney ML (1989) Interaction between
oral ciprofloxacin and caffeine in normal volunteers. Antimicrob Agents
Chemother 33:474--478
Helsing E (1992) Vitamins. In: Dukes MNG (ed) Side effect of drugs, 11 edn. Elsevier,
Amsterdam, pp 959-976
Hesseling PB, Klopper JF, van Heerden PD (1979) Die effek van Rooibostee op
ysterabsorpsie. S Afr Med J 55:631-632
Hirsch SR (1979) Precipitation of antipsychotic drug in interactions with coffee or tea.
Lancet 11:1130-1131
Hogan RP (1983) Hemorrhagic diathesis caused by drinking a herbal tea. JAMA
249:2679-2680
Homma M, Oka K, Ikeshima K, Takahashi N, Yamamoto S, Niitsuma T, Hoh H
(1992a) Different effects of three Kampo-remedies which compromise of saiko on
prednisolone pharmacokinetics. 5th World Conference on Clinical Pharmacology
and Therapeutics, July 26-31, Yokohama
Homma M, OkaK, Yamada T, Niitsuma T, Itoh H, Takahashi N (1992b) A strategy for
discovering biologically active compoounds with high probability in traditional
Chinese herb remedies: an application of Saiboku-to in bronchial asthma. Anal
Biochem 203:179-187
Homma M, Oka K, Niitsuma T, Hoh H (1993) Pharmacokinetic evaluation of tradi-
tional Chinese herbal remedies. Lancet 341:1595
Homma M, Oka K, Niitsuma T, Hoh H (1994) A novel 11 beta-hydroxysteroid dehy-
drogenase inhibitor contained in Saiboku-To, a herbal remedy for steeroid-depen-
dent bronchial asthma. J Pharm Pharmacol 46:305-309
Hoppe HA (1975) Angiospermen, 8 edn. Gruyter, Berlin, pp 91-92, 435, 519-520, 700-
701 (Drogenkunde, vol 1)
Huupponen R, Karhuvaara S, Seppala P (1985) Effect of guar gum on glipizide absorp-
tion in man. Eur J Clin PharmacoI28:717-719
Ingram CG (1962) Some pharamcologic actions of yohimbine and chlorpromazine in
man. Clin Pharmacol Ther 3:345-352
Ito K (1993) Effects of Sairei-to on nephrotic syndrome. J Tokyo Womens Med Coll
63:464-468
Joarder ZH, Ogawa T, Yorioka N, Yamakido M (1991) Studies on the effectiveness of
Sairei-to on puromycin aminonucleoside nephrosis in rats. Hiroshima J Med Sci
40:127-135
Johri RK, Zutshi U (1992) An Ayurvedic formulation "Trikatu" and its constituents.
J Ethnopharmacol 37:85-91
Jones BD, Runikis AM (1987) Interaction with ginseng. J Clin Psychopharmacol
7:201-202
Jori A, Bianchetti A, Prestini PE (1969) Effect of essential oils on drug metabolism.
Biochem Pharmacol 18:2081-2085
350 P.A.G.M. DE SMET and P.F. D'ARCY
Joseph AM, Biggs T, Garr M, Singh J, Lederle FA (1991) Stealth steroids. N Engl J
Med 324:62
Karunanithy R, Sumita KP (1991) Undeclared drugs in Chinese antirheumatoid medi-
cine. Int J Pharm Pract 1:117-119
Kato H, Nakanishi K, Kanoaka M (1984) The enzyme immunoassay of glycyrrhizin and
glycyrrhetinic acid and its clinical application. Minophagen Med Rev 15 [Suppl]:3-
11
Kempin SJ (1983) Warfarin resistance caused by broccoli. N Engl J Med 308:1229-1230
Khanna P, Jain SC, Panagariya A, Dixit VP (1981) Hypoglycemic activity of polypep-
tide-P from plant source. J Nat Prod 44:648-655
Khin-Maung-U, Myo-Khin, Nyunt-Nyunt-Wai, Aye-Kyaw, Tin-U (1985) Clinical trial
of berberine in acute watery diarrhoea. Br Med J 291:1601-1605
Kimura K, Nanba S, Tojo A, Matsuoka H, Sugimoto T (1990) Effects of Sairei-to
on the relapse of steroid-dependent nephrotic syndrome. Am J Clin Med 18:45-
50
Koren G, Boichis H, Keren G (1982) Effect of tea on the absorption of pharmacologic
doses of an oral iron preparation. Isr J Med Sci 18:547
Kulhanek F, Linde OK (1981) Coffee and tea influence pharmacokinetics of
antipsychotic drugs. Lancet 11:359-360
Kulhanek F, Linde OK, Meisenberg G (1979) Precipitation of antipsychotic drugs in
interaction with coffee and tea. Lancet 11:1130
Kunin CM, Finland M (1961) Clinical pharmacology of the tetracycline antibiotics. Clin
Pharmacol Ther 2:51-69
Kwan T, Paiusco AD, Kohl L (1992) Digitalis toxicity caused by toad venom. Chest
102:949-950
Lacomblez L, Bensimon G, Isnard F, Diquet B, Lecrubier Y, Puech AJ (1989) Effect
of yohimbine on blood pressure in patients with depression and orthostatic
hypotension induced by clomipramine. Clin Pharmacol Ther 45:241-251
La Piana Simonsen L (1989) Top 200 drugs of 1988. Pharmacy Times 40
Lasswell WL Jr, Weber SS, Wilkins JM (1984) In vitro interaction of neuroleptics and
tricyclic antidepressants with coffee, tea and gallotanic acid. J Pharm Sci 73:1056-
1058
Leatherdale BA, Panesar RK, Sing G, Atkins TW, Bailey CJ, Bignell AH (1981)
Improvement in glucose tolerance due to Momordica charantia (karela). Br Med
J 282:1823-1824
Lever PG, Hague JR (1964) Observations on phenothiazine concentrates and diluting
agents. Am J Psychiatry 120:1000-1002
Lewis WH, Elvin-Lewis MPF (1977) Medical botany. Plants affecting man's health.
Wiley, New York, p 192
Lin C-S, Lin M-C, Chen K-S, Ho C-C, Tsai S-R, Ho C-S, Shieh W-H (1989) A digoxin-
like immunoreactive substance and atrioventricular block induced by a Chinese
medicine "Kyushin". Jpn Circ J 53:1077-1080
Loeper J, Descatoire V, Letteron P, Moulis C, Degott C, Dansette P, Fau D, Pessayre
D (1994) Hepatotoxicity of germander in mice. Gastroenterology 106:464-472
Mahr G, Soergel F, Granneman GR, Kinzig M, Muth P, Patterson K, Fuhr U, Nickel
P, Stephan U (1992) Effects of temafloxacin and ciprofloxacin on the pharmacoki-
netics of caffeine. Clin Pharmacokin 22 [Suppl 1]:90-97
McIvor ME, Cummings CC, Mendeloff AI (1985) Long-term ingestion of guar gum is
not toxic in patients with noninsulin-dependent diabetes mellitus. Am J Clin Nutr
41:891-894
Medical Letter (1971) Vitamin C - were the trials well conducted and are large doses
safe? Med Lett Drugs Ther 13:48
Merkel U, Sigusch H, Hoffmann A (1994) Grapefruit juice inhibits 7-hydroxylation of
coumarin in healthy volunteers. Eur J Clin PharmacoI46:175-177
Monder C, Stewart PM, Lakshmi V, Valentino R, Burt D, Edward CRW (1989)
Licorice inhibits corticosteroid II-dehydrogenase of rat kidney and liver: in vitro
and in vivo studies. Endocrinology 125:1046-1053
Drug Interactions with Herbal and Other Non-orthodox Remedies 351
Soons PA, Vogels BAPM, Roosemalen CM, Schoemaker HC, Uchida E, Edgar B,
Lundahl J, Cohen AF, Breimer DD (1991) Grapefruit juice and cimetidine inhibit
stereos elective metabolism of nitrendipine 'in humans. Clin Pharmacol Ther
50:394-403
Stewart DE (1992) High-fiber diet and serum tricyclic antidepressant levels. J Clin
Psychopharmacol 12:438-440
Stinson JC, O'Gharabhain F, Adebayo G, Chambers PL, Feely J (1993) Pesticide-
contaminated cucumber. Lancet 341:64
Stockley IH (1984) Warfarin resistance caused by over-the-counter drugs, food supple-
ments and vegetables. Pharm Int 5:165-167
Stockley IH (1991) Drug interactions. A source book of adverse interactions, their
mechanisms, clinical importance and management, 2nd edn. Blackwell, Oxford
Sunter WH (1991) Warfarin and garlic. Pharm J 246:722
Tamura Y, Nishikawa T, Yamada K, Yamamoto M, Kumagai A (1979) Effects of
glycyrrhetinic acid and its derivatives on ~4-5a- and 5f3-reductase in rat liver.
Arzneimittelforschung 26:647-649
Taylor RFH, AI-Jarad N, John LME, Conroy DM, Barnes NC (1992) Betel-nut chew-
ing and asthma. Lancet 339:1134-1136
Teuscher E, Lindequist U (1988) Biogene Gifte. Biologie-Chemie-Pharmakologie.
Academie, Berlin, pp 228-229
Todd P A, Benfield P, Goa KL (1990) Guar gum. A review of its pharmacological
properties and use as a dietary adjunct in hypercholesteraemia. Drugs 39:917-928
Tsumura A (1991) Kampo - how the Japanese updated traditional herbal medicine.
Japan Publications, Tokyo
Udall JA, Krock LB (1968) A modified method of anticoagulant therapy. Curr Ther
Res 10:207-211
Uusitupa M, Siitonen 0, Savolainen K, Silvasti M, Penttila I, Parviainen M (1989)
Metabolic and nutritional effects of long-term use of guar gum in the treatment
of noninsulin-dependent diabetes of poor metabolic control. Am J Clin Nutr
49:345-351
Vesell ES, Shively CA, Passananti GT (1979) Failure of cola nut chewing to alter
antipyrine disposition in normal male subjects from a small town in South Central
Pennsylvania. Clin Pharmacol Ther 26:287-293
Walker FB (1984) Myocardial infarction after diet-induced warfarin resistance. Arch
Intern Med 144:2089-2090
Watson AJM, Pegg M, Green JRB (1984) Enteral feeds may antagonise warfarin. Br
Med J 288:557
Wesley-Hadzija B (1971) A note on the influence of diet in West Africa on urinary pH
and excretion of amphetamine in man. J Pharm Pharmacol 23:366-368
Yoneyama Y, Usukura Y, Arai H, Hasegawa R, Haranaka R, Nakagawa S (1990)
Effects of Sairei-to on glutathione metabolism in alcohol-treated mice. Nihon
Univ J Med 32:379-387
Subject Index
cephalexin 14, 31, 56, 183, 194, 296, 316 closapine 219
cephalothin 183, 194, 310, 311 cocaine 216,217,219,220,221
cephradine 29, 31, 182, 183 codeine 296,316
CGP 43371 80,87,88 cola nut 336
CHAP 5 245 colchicine 17, 296, 327
charcoal 14,16,21,313 colestipol 14
chelate formation 19 colistine 296
chinese toad venom 329 Committee for Proprietary Medicinal
chloral hydrate 26, 130, 296 Products of the European
chlorambucil 296 Community 345
chloramphenicol 160,162,296,317 contact lenses, drug interactions with
chlorazepate, dipotassium 256 265
chlorcyclizine 161 Convallaria majalis 340
chlordiazepoxide 26, 138, 263, 286, 288, coumarin 327,332,344
296 creatinine 310, 311, 317
chlorinated hydrocarbons 160 analysis 317
chlormethiazole 264, 286, 296 clearance 190, 202
chlormezanone 296 Crohn's disease 126
2-chloro-2'-deoxyadenosine 52, 56 cyanide 309
chloroform 296 cyclacillin 31
chloroquine 6,226,266,296,312,332 cyclamate sodium 15
chlorothiazide 14,25,203 cyclizine 296
chlorpheniramine 271,296 cyclophosphamide 37, 162, 164,245,
chlorpromazine 14,152,220,223,226, 286, 296
255,284,296,298,335,343 cyclopropane 296
chlorpropamide 206,296,341 cyclosporine 6, 34, 35, 81, 89, 100, 105,
chlortetracycline 14, 19 153,161,162,163,227,246,263,265
chlorthalidone 286 cyclosporine A 263
chlorzoxazone 153,312 CYPIA 152
cholestyramine 14,17,20 CYPIAI 153
cholesterol 155 CYPIA2 153, 157, 334
cholecystokinin 8, 87 CYP2A6 153, 157
choline 222 CYP2A6 v 157
cholinergic receptors 223 CYP2A7 153
synapse 222 CYP2B 152
cicaprost 52 CYP2B6 153
cimetidine 4, 100, 105, 160, 162, 164, CYP2B7 153
176, 188, 189, 190, 191, 192, 193, CYP2C 152
194,240,246,283,286,296,312 CYP2C9 153
cinoxacin 182 CYP2CI0 153
ciprofioxacin 14,27, 52, 57, 61, 160, CYP2C17 153
161,162,338 CYP2C18 153
circadian rhythm 76,77 CYP2C19 153
cisapride 223 CYP2D 152
cisplatin 245,251, 271 CYP2D6 153, 339
citalopram 220,221 CYP2D7P 153
CL 275838 65 CYP2D8P 153
clarithromycin 80, 81, 89, 241, 242 CYP2E 152
clindamycin 14,134 CYP2El 153
clobazam 288, 296 CYP2F 152
clofazimine 240, 246, 313 CYP2Fl 153
clofibrate 160, 179, 296 CYP3A 34, 35, 152, 154
clomipramine 220, 256 CYP3A3 153, 154
clonazepam 240, 296 CYP3A4 49,153,154,157,332
clonidine 216,217,218,296 CYP3A5 153, 157
clorgyline 3 CYP3A7 153
356 Subject Index